Download as pdf or txt
Download as pdf or txt
You are on page 1of 95

REVIEW ARTICLE

CARDIAC TRANSMEMBRANE ION CHANNELS


AND ACTION POTENTIALS: CELLULAR
PHYSIOLOGY AND ARRHYTHMOGENIC
BEHAVIOR
Remodeling Genetic mutations,
AUTHORS
Channel/protein expression
 , Jakub Tomek, Norbert Nagy,
(disease, chronic drug) polymorphisms
András Varro
Acute drug effects
Acute diseases
Transmembrane ion channel/pump function Hormones Gender  Virág, Elisa Passini, Blanca Rodriguez,
Lászlo
(e.g. ischemia)
Transmembrane cellular voltage change
X+
Amp
István Baczko
X+
Single cell action potential X+ +
Plasma electrolyte X+ X
alterations
Tissue action potential + impulse conduction Amp
Single cell action potential + electrotonic interaction Ce

Whole heart summated electrical signal


CORRESPONDENCE
Electrocardiogram – clinical diagnostic tool
varro.andras@med.u-szeged.hu
TdP
Arrhythmia
AF Substrate Trigger
VF

Impulse Repolarization/ERP Triggered activity


Enhanced normal
automaticity
KEY WORDS
conduction heterogeneity
DAD EAD SAN Purkinje fiber
action potential; arrhythmia; heart; ion channels;
remodeling

Pathological settings

Plasma electrolyte Myocardial Heart HCM/ Diabetes Inherited/genetic Drugs/Dietary ingredients


imbalance ischemia failure Athletes heart mellitus abnormalities

CLINICAL HIGHLIGHTS
1. Cardiac arrhythmias are major causes of mortality. They most often arise from pathological changes in the electro-
physiological properties of myocardial cells. First, this review summarizes the physiology of cardiac action poten-
tials, their regional and species differences, the underlying transmembrane ionic currents, and transporters, with
special focus on their human relevance.
2. Progress in computer modeling and vast quantities of experimental data made computerized replication of the action
potential, impulse conduction, and simulation of cardiac electrophysiology possible. Current computer models offer
improved observability and controllability via utilization of different modeling scales and can assist future individualized
anti-arrhythmic therapy as well as drug electrophysiological safety assessment.
3. A number of diseases evoke changes in the configuration of the action potential caused by altered function and/or den-
sities of transmembrane ion channels and transporters, collectively termed electrical remodeling. Initially, these altera-
tions are often compensatory; however, remodeling significantly contributes to increased arrhythmia susceptibility by
impairing impulse generation, conduction, and myocardial refractoriness in these clinical settings. Electrical remodeling
in atrial fibrillation, heart failure, hypertrophic cardiomyopathy, myocardial infarction, and advanced age are discussed.
Better understanding of the cellular basis of cardiac electrophysiology, electrical remodeling, and mechanisms of
arrhythmias has important implications for future clinical therapeutic strategies.

VARRÓ ET AL., 2021, Physiol Rev 101: 1083–1176


October 29, 2020; Copyright © 2021 The Authors. Licensed under Creative Commons Attribution CC-Y 4.0. Published by the American
Physiological Society
https://doi.org/10.1152/physrev.00024.2019
Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
Physiol Rev 101: 1083–1176, 2021
First published October 29, 2020; doi:10.1152/physrev.00024.2019

REVIEW ARTICLE

CARDIAC TRANSMEMBRANE ION CHANNELS


AND ACTION POTENTIALS: CELLULAR
PHYSIOLOGY AND ARRHYTHMOGENIC
BEHAVIOR
,1,2 Jakub Tomek,3 Norbert Nagy,1,2 Lászlo
András Varro  Virág,1 Elisa Passini,3 Blanca Rodriguez3, and István Baczko
1
1
Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary; 2MTA-SZTE
Cardiovascular Pharmacology Research Group, Hungarian Academy of Sciences, Szeged, Hungary; and 3Department of Computer
Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom

Abstract
Cardiac arrhythmias are among the leading causes of mortality. They often arise from alterations in the electro-
physiological properties of cardiac cells and their underlying ionic mechanisms. It is therefore critical to further
unravel the pathophysiology of the ionic basis of human cardiac electrophysiology in health and disease. In the
first part of this review, current knowledge on the differences in ion channel expression and properties of the
ionic processes that determine the morphology and properties of cardiac action potentials and calcium dynam-
ics from cardiomyocytes in different regions of the heart are described. Then the cellular mechanisms promoting
arrhythmias in congenital or acquired conditions of ion channel function (electrical remodeling) are discussed.
The focus is on human-relevant findings obtained with clinical, experimental, and computational studies, given
that interspecies differences make the extrapolation from animal experiments to human clinical settings difficult.
Deepening the understanding of the diverse pathophysiology of human cellular electrophysiology will help in
developing novel and effective antiarrhythmic strategies for specific subpopulations and disease conditions.
action potential; arrhythmia; heart; ion channels; remodeling

1. INTRODUCTION 1083 CLINICAL HIGHLIGHTS


2. CARDIAC ACTION POTENTIAL 1086
1. Cardiac arrhythmias are major causes of mortality. They most
3. TRANSMEMBRANE ION CHANNELS AND... 1087 often arise from pathological changes in the electrophysiolog-
4. TISSUE-SPECIFIC ACTION POTENTIALS 1107 ical properties of myocardial cells. First, this review summa-
5. COMPUTER SIMULATIONS OF ACTION... 1117 rizes the physiology of cardiac action potentials, their regional
6. CELLULAR ARRHYTHMIA MECHANISMS 1120 and species differences, the underlying transmembrane ionic
7. ION CHANNEL AND ACTION POTENTIAL... 1126 currents, and transporters, with special focus on their human
relevance.
8. INHERITED CONDITIONS ASSOCIATED... 1132
2. Progress in computer modeling and vast quantities of experimen-
9. OTHER FACTORS INFLUENCING... 1134 tal data made computerized replication of the action potential,
10. CONCLUSIONS 1136 impulse conduction, and simulation of cardiac electrophysiology
possible. Current computer models offer improved observability
and controllability via utilization of different modeling scales and
can assist future individualized anti-arrhythmic therapy as well as
drug electrophysiological safety assessment.
1. INTRODUCTION 3. A number of diseases evoke changes in the configuration of the
action potential caused by altered function and/or densities of
transmembrane ion channels and transporters, collectively
The heart is a mechanical pump with the vital role of termed electrical remodeling. Initially, these alterations are often
supplying blood to other organs. In humans, it contracts compensatory; however, remodeling significantly contributes to
and relaxes in a regular fashion 60 times per minute. If increased arrhythmia susceptibility by impairing impulse genera-
the regular heartbeat is interrupted for more than a cou- tion, conduction, and myocardial refractoriness in these clinical
settings. Electrical remodeling in atrial fibrillation, heart failure,
ple of minutes, the lack of oxygen supply causes irre- hypertrophic cardiomyopathy, myocardial infarction, and
versible damage to vital organs, including the heart advanced age are discussed. Better understanding of the cellu-
itself, potentially causing sudden cardiac death (SCD). lar basis of cardiac electrophysiology, electrical remodeling, and
mechanisms of arrhythmias has important implications for future
Cardiac contractions, the most important function of the
clinical therapeutic strategies.
heart, are initiated by a bioelectrical signal, the action

0031-9333/21 Copyright © 2021 The Authors. Licensed under Creative Commons Attribution CC-BY 4.0. 1083
Published by the American Physiological Society.
Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

potential (AP) (1), via a process called excitation-contrac- heart as measured on the body surface. The ECG is
tion coupling (2). The action potential originates in the determined by many variables, including the function of
sinus node cells, propagates through the whole heart transmembrane ion channels and transporters in the dif-
via an active electrophysiological process called impulse ferent heart cells and the consequent changes in the
conduction, and can be measured as the electrical membrane potential (FIGURE 1).
potential difference between the intra- and extracellular The P wave corresponds to the activation (depolari-
space. The stimulus spreads through both atria, causing zation) and early repolarization of the atrial cells. The
their contraction. The next stage of propagation is the QRS complex reflects the time course of the depolari-
atrioventricular (AV) node, which passes the signal to zation of the ventricles caused mainly by the activation
the ventricles with a slight delay to provide enough time of the fast sodium channels. The PQ segment mainly
for the atria to contract. From the AV node, the bundle indicates the impulse conduction from the atria to the
of His conducts the stimulus along the septal wall ventricles. The PQ segment also contains the HQ inter-
to the subendocardial Purkinje fibers, which then stimu- val, which reflects fast propagation due to the function
late the ventricles, allowing their synchronized contrac- of the fast sodium current (INa). In addition, cell-to-cell
tion. The action potential is determined by the opening coupling is low in the AV node (8), which makes
and closing of various complex transmembrane pro- impulse propagation through the AV node relatively
teins, which consist of ion channels and transporters, unsafe. The isoelectric ST segment reflects the plateau
i.e., pumps or exchangers (3–5). Disturbances of action phase of the ventricular action potentials. In this phase,
potential generation and/or conduction can lead to membrane potential hardly changes at the cellular
changes in the regular heart rhythm called arrhythmias level because of the fine balance of opening/closing of
(6, 7). These disturbances can impair contraction to such different ion channels. The configuration of the T wave
a degree that thromboembolic stroke of atrial origin or shows the repolarization time course of the ventricles,
sudden cardiac death may eventually occur. Therefore, and it reflects the balance between the slowly activat-
understanding the function and regulation of transmem- ing repolarizing potassium and chloride currents and
brane ion channels and transporters, as well as their the depolarizing steady-state, so-called “window” so-
impact on the cardiac action potential, is essential to dium (9) and “window” calcium (10) and the slowly
understand arrhythmia mechanisms and treat life-threat- decaying, often called “late,” sodium (INaLate) and
ening cardiac arrhythmias. Arrhythmias are usually diag- slowly inactivating calcium currents (11–15). Analysis of
nosed based on the analysis of electrocardiogram (ECG) the PP intervals yields important information regarding
recordings, which represent the electrical activity of the heart rate and its regularity.

FIGURE 1. Regional differences in cardiac


action potential configurations. Schematic cross
section of the heart depicting the corresponding
action potential configuration from different
regions of the heart indicated by arrows.
Color-coded sections on the action potentials
refer to the corresponding sections on the
schematic electrocardiogram (ECG). AVN,
atrioventricular node; Endo, endocardial; Epi,
epicardial; Mid, midmyocardial; SAN, sinoa-
trial node.

1084 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

FIGURE 2. A, top: an area of branching cardiac tissue, providing separate paths for impulse propagation from proximal (1) to distal (2–4) directions. The
separate paths for impulse conduction can be variable, as branching Purkinje fiber-ventricular junctions, ventricular muscle segments with nonconducting
fibrosis, scar, or infarcted tissue in the core. If the tissues in these paths are healthy, the impulse conduction is fast and, because of the relatively long effec-
tive refractory period (ERP) in cardiac cells, impulses would collide at site 4 and would propagate only into the distal directions. Bottom: an area of depolar-
ized myocardium at site 3, due to asymmetric severe local myocardial damage, imposing conduction block from the anterograde direction. However, in
the case that the impulse travels from site 4 in the retrograde direction, it can propagate back very slowly into this damaged area, and if its propagation is
slow enough to outlast the ERP in front the impulse can reexcite the proximal tissue. Then, this impulse would propagate toward both sites 1 and 2, estab-
lishing a circus movement and reentry arrhythmia. B: example of a classic mechanism by which a premature ventricular complex (PVC) initiates reentry in
the fibrotic border zone of an infarct, due to slow conduction and dispersion of refractoriness. Top: a PVC occurring 250 ms after the previous beat arrives
too early to propagate through the upper myocyte strand with a long ERP of 275 ms, but it propagates successfully (red arrows) through the lower strand
with a shorter ERP of 225 ms (entry site). The impulse propagates slowly (slow CV), eventually reaching the upper strand from the opposite direction.
Bottom: if the total conduction time is >275 ms, the interface of the upper strand with normal tissue (exit site) has recovered excitability, and the impulse
can then propagate through the region of prior conduction block, thus initiating reentry. Dispersion of refractoriness is caused by electrical remodeling.
The slow propagation is due to zig-zag conduction through the myocyte strands as well as gap junction remodeling. (Reproduced from Ref. 17 with permis-
sion.) C: schematic illustration of functional reentry mechanism without a well-defined anatomical obstacle. The arrhythmia substrate is represented by arti-
ficially enhanced action potential duration differences. In normal circumstances, impulses originating from the sinus node (black arrows) use physiological
pathways to propagate through atrial and ventricular tissue and the conduction system. An early ectopic impulse (trigger, red arrow) can only propagate
via pathways where the tissue is not depolarized, and consequently its refractoriness is over, whereas the conduction is blocked in directions where the
tissue is not fully repolarized and cells are still in the refractory state. Thus the abnormal impulse can travel in a zig-zag direction through reentry paths cre-
ated by heterogeneous repolarization and conduction. The dispersion of repolarization creates a time window called the vulnerable period, where extra
stimuli could elicit the reentry arrhythmia. However, outside this window extra stimuli would only cause a single or multiple relatively harmless extrasys-
toles. CV, conduction velocity; ES, extrasystole; SR, sinus rhythm. Reproduced from Ref. 18 with permission.

Cardiac arrhythmia mechanisms are still the sub- widely accepted that there is not a single mechanism
ject of intensive research. Because of the large vari- to explain how arrhythmias originate. Therefore,
ability in appearances, types (e.g., bradycardia and patients are often treated with little knowledge
different types of tachycardia), locations (supraven- regarding the mechanisms and/or causes of the
tricular or ventricular), and underlying diseases, it is arrhythmia.

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1085


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

The majority of cardiac arrhythmias are the result of of inwardly rectifying K1 currents and can be roughly
an enhanced proarrhythmic substrate combined with a estimated by the Nernst equation from the uneven distri-
trigger (16). Enhanced heterogeneity of repolarization bution of mainly K1 ions across the cell membrane. The
and impaired impulse conduction represent typical ar- electrogenic ATP-dependent Na1-K1 pump also con-
rhythmia substrates (conditions that are prerequisites for tributes to the RMP, by exporting 3 Na1 and importing 2
arrhythmia development) for severe tachyarrhythmia. K1 (35–38). In healthy conditions, the duration of the
Impairment of impulse conduction can be caused by an- action potential (APD) determines the effective refrac-
atomical (FIGURE 2, A and B) (17) or functional (FIGURE tory period (ERP), defined as the shortest time interval
2C) (18) alterations. The process was described long needed before a new stimulus, or an early extrasystole,
ago, first in the early twentieth century (19). Impulse con- can elicit another action potential. The relationship
duction critically depends on the density and kinetics of between APD and ERP can be disrupted in pathological
inward transmembrane ionic currents. Depolarization of conditions, for example, in hyperkalemia, resulting in
the resting membrane potential (RMP), for example, postrepolarization refractoriness (39).
reduces sodium and calcium inward currents and In the context of the cardiac action potential, two
strongly influences their kinetic properties. This can thus aspects should be emphasized. First, there is no such
slow impulse conduction and cause unidirectional or uniform entity as “the cardiac action potential,” since its
bidirectional conduction block, and potentially reentry, shape, i.e., the time course of the transmembrane poten-
underpinning a wide range of cardiac arrhythmias. tial changes, differs in the various regions of the heart
As mentioned above, reentrant arrhythmias can be (FIGURE 1), and therefore different action potentials
caused by functional causes too, without an anatomically should be considered and discussed separately.
well-defined myocardial damage (FIGURE 2C). This form of Second, there are significant interspecies differences
reentry is more complex and could involve both impulse (40), even when action potentials are recorded from sim-
conduction and repolarization heterogeneities (arrhythmia ilar regions of the heart. This is an important, and often
substrate) as well as enhanced normal or abnormal automa- overlooked, issue, since many experimental results
ticity (trigger). have been obtained in small rodents, recently particu-
Reentrant arrhythmias are often initiated by an extrasys- larly in transgenic mice.
tole formed anywhere in the heart, acting as an arrhythmia In general, the cardiac action potential is divided into
trigger (20, 21). Both arrhythmia substrate and trigger, like five distinct phases (FIGURE 1). Phase 0 is the fast depo-
an extrasystole [premature ventricular complex (PVC)], can larization due to an abrupt increase in sodium influx, and
be promoted by pathological cardiac conditions (FIGURE it is characterized by the upstroke velocity and can result
2), e.g., myocardial ischemia, heart failure (7), and genetic in an overshoot, i.e., the rapid change of potential from
diseases (22), or by adverse drug reactions (23). General the negative RMP to positive voltage values, reaching a
arrhythmia mechanisms include various cellular aspects, peak of up to 130 to 140 mV. The overshoot is fol-
e.g., transmembrane ionic currents, transporters, action lowed by a return to negative values, in a process called
potential properties, and automaticity, which are the sub- repolarization, which includes phases 1, 2, and 3. Phase
jects of this review. However, arrhythmia mechanisms at 1 is characterized by a transient and relatively fast repo-
the whole heart level are more complex, since they are larization brought about by a decrease in sodium influx
also determined by anatomical and structural properties, and a transient increase in potassium efflux and chloride
impulse conduction, and intercellular communication influx. Phase 2 consists of a long-lasting plateau, still at
between myocardial and nonmyocardial cells, like fibro- depolarized voltage, during which the membrane poten-
blasts. These factors are beyond the scope of the present tial remains almost constant or decreases slowly,
work, and the interested reader is referred to other reviews caused by a small net transmembrane current carried by
(22, 24–34). simultaneous calcium (and some sodium) influx and po-
tassium efflux. Phase 3 represents the large repolariza-
tion toward the diastolic potential, mostly due to
2. CARDIAC ACTION POTENTIAL increased potassium efflux and decreased calcium and
sodium influx. Phase 4 represents the resting membrane
The cardiac action potential is a transmembrane poten- potential in diastole in working myocytes and the spon-
tial change, with an amplitude ranging between 60 and taneous depolarization in pacemaker cells. In cardiac
120 mV. It starts from a negative value, i.e., the resting myocytes that do not beat spontaneously the voltage
membrane potential (RMP) in working myocardial cells remains stable at the RMP, whereas in cells exhibiting
or maximal diastolic potential in spontaneously beating automaticity the potential gradually changes toward the
cells (1), ranging from 95 to 40 mV. As in other excita- positive values, in a process called spontaneous dia-
ble cells, the RMP is mainly defined by the conductance stolic depolarization. When the threshold potential is

1086 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

reached, a new spontaneous action potential is gener- opening/closing be-havior of ion channels has a more
ated, with a certain cycle length. profound effect on membrane potential than in well-
There are four common methods to record cardiac coupled tissue preparations (50). In addition, in multicel-
action potentials: lular preparations part of the ionic currents are utilized to
1) Weidmann and Coraboeuf were the first to record car- depolarize neighboring cells during impulse propaga-
diac action potentials in dog ventricular muscle (41) and tion, and this can considerably reduce the action poten-
later in dog Purkinje fibers, using the sharp glass capil- tial peak compared with single cells (51). Hence, action
lary-based microelectrode technique (42). The classical potential measurements in single isolated myocytes
cardiac cellular electrophysiological knowledge gained obtained with the patch-clamp technique should be
by using this technique was elegantly summarized in interpreted with caution and not directly extrapolated to
an early monograph entitled Electrophysiology of the intact tissue.
Heart by Hoffman and Cranefield (1), published in 1960, 4) The latest approach to recording cardiac action poten-
which is still useful today. This technique is still consid- tials is the optical mapping technique, which uses volt-
ered one of the best for accurate cardiac action poten- age-sensitive dyes and allows simultaneous recordings
tial recordings and can be used for both single-cell and from multiple sites (52, 53). This technique is also excel-
tissue recordings. Its major advantages include 1) the lent for dynamic studies and for investigations of arrhyth-
ability to accurately record very fast voltage changes mia mechanisms (54, 55). Disadvantages of this method
and 2) because of the very fine tip of the pipette, very include the difficulty of calibration to millivolts, phototox-
little diffusion takes place out of the pipette solution, icity, photodegradation, and photon scattering effects
having negligible effects on the intracellular milieu. (56). Also, the application of excitation-contraction
However, since this technique has some limitations uncoupling compounds, e.g., blebbistatin, is necessary
(e.g., difficulties in maintaining a stable impalement for to avoid motion artifacts (57). These compounds may
extended periods of time), other methods have also interfere with the experiments, since, e.g., blebbistatin
been developed and used widely. was reported to elicit anomalous electrical activities (58)
2) In intact hearts, in in vivo animal experiments, or in clini- and prolongation of action potential duration (59), and in-
cal studies, where the microelectrode technique is diffi- hibition of contraction will also decrease metabolic rate
cult to apply, monophasic action potential recording at the concentrations needed for motion artifact red-
can also be used, with either a suction electrode (43, uction.
44) or a Franz catheter (45). With this technique,
recordings can be easily performed from multiple sites
simultaneously, and impalements/attachments are not 3. TRANSMEMBRANE ION CHANNELS AND
lost because of vigorous contractions, even in in vivo TRANSPORTERS IN THE HEART
or ex vivo conditions. However, rapid voltage changes
or action potential amplitudes and shapes cannot be The cardiac action potential is the voltage change
determined accurately. caused by ions flowing through transmembrane ion
3) Since the introduction of the patch clamp by Neher and channels, via their dynamic and simultaneous opening
Sakmann (46–48), the whole cell configuration of this and closing (11, 60). Therefore, before addressing differ-
technique has been widely used. In the current-clamp ent regional action potential patterns, we describe the
mode, it can record action potentials from isolated myo- various transmembrane ion channels that have been
cytes. Despite its widespread use, this technique has im- reported to operate in the heart cells.
portant limitations that should be emphasized. First,
Transmembrane ionic currents in the heart are usu-
measurements are performed in single isolated myo-
ally measured with the patch-clamp technique in
cytes or, occasionally, cell pairs, and it is uncertain how,
enzymatically isolated myocytes. This allows record-
and to what degree, different ion channels are influ-
ing and analysis of unitary currents through single
enced by individual enzymatic digestion during the isola-
tion procedure (49). Therefore, even if the recordings ion channels or all channels on the sarcolemma.
show single-cell action potentials with a normal shape, Before the introduction of the patch clamp, trans-
the function of the finely regulated ion channels can be membrane current recordings were less accurate,
drastically altered from their original condition. Also, the because of the lack of proper voltage control of the
cell is dialyzed with the pipette contents, and its intracel- preparation, and fast current changes and gating
lular composition will change. When carefully and delib- kinetics were impossible to determine accurately
erately applied, however, this point can also be (61). Therefore, much of the knowledge gained
considered an advantage, as it allows control of the intra- before the introduction of the patch-clamp technique
cellular milieu. It should also be emphasized that single has had to be reevaluated, and some currents have
isolated myocytes are devoid of electrotonic interactions been renamed. To the interested reader, we recom-
from neighboring cells. Therefore, the stochastic mend an excellent monograph by Denis Noble, The

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1087


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

FIGURE 3. Schematic illustration of voltage-gated sodium (Nav) and potassium (Kv) channels. Top: topological models of Nav1.5 channel subunits and
their molecular assembly. Four domains (I–IV) of Nav a-subunits contribute to individual Nav channel formation. Middle: the activated and inactivated
Nav channel configurations. Bottom: the transmembrane topologies of Kv, inward rectifier (Kir), and two-pore domain (K2P) potassium channel subunits.

Initiation of the Heartbeat (62), which deductively most transmembrane ion channels consist of multiple
and briefly summarizes our knowledge before the subunits: a pore-forming a and modulatory accessory
use of the patch-clamp technique. subunits, which can modify channel gating and serve
It is important to note that a given ionic current, meas- as possible drug binding or phosphorylation sites (3, 5,
ured with the patch clamp, does not correspond to a 67, 68).
unique ion channel. Certain transmembrane ionic cur-
rents can be conducted by several different ionic chan- 3.1. The Fast Inward Sodium Current
nels (3, 5), e.g., inward rectifier potassium current (IK1) is
carried by inward rectifier potassium (Kir)2.1, Kir2.2, The fast inward sodium current (INa) is the most impor-
Kir2.3, and Kir2.4 channels (5, 63) and tandem of pore tant current for impulse conduction in cardiomyocytes,
domains in a weak inward rectifying potassium channel with a diastolic potential more negative than 60 mV
(TWIK)-related acid-sensitive potassium (TASK) channels (69), and is therefore particularly important in atrial, ven-
(64), whereas transient outward current (Ito) is carried by tricular, and Purkinje fiber myocytes. This current is re-
voltage-gated potassium (Kv)4.3, Kv4.2, and Kv1.4 chan- sponsible for the influx of Na1 during phase 0 of the
nels. In addition, certain channels like human ether-à- action potential (FIGURE 1 and FIGURE 4), and it is con-
go-go-related gene (hERG) have multiple isoforms ducted by voltage-gated sodium (Nav)1.5 channels,
(hERG 1a and 1b) (65) with different gating kinetics and encoded by the gene SCN5A coassembled with b1–4
drug sensitivities (66). This offers the possibility of phar- (SCN1–4B)-subunits (70). The high-molecular-weight
macologically modulating specific channel isoforms pore-forming a-subunit contains four repeat domains (la-
without interfering with others, and thus avoiding unde- beled I–IV); each domain consists of six transmembrane
sirable side effects. segments (S1–S6), and the S4 segment is responsible
Recent advances in genetics and molecular biology for voltage sensing (FIGURE 3). Special extracellular
have made it possible to further elucidate the structure regions (P loops) between the S5 and S6 segments of
of ion channels (FIGURE 3). It is widely known that the four domains form the structure that is responsible

1088 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

channels can recover from inactivation with a time con-


stant of 2–20 ms at negative voltages (69, 73, 74). The
exact kinetics of their inactivation and recovery are com-
plex and still not fully understood. So far, multiple inacti-
vation kinetics and recovery kinetics from inactivation
have been described (12, 69, 75–77).
In addition to this fast component, slow inactivation,
occurring over hundreds of milliseconds, has been
described (12, 13, 78) and attributed to late openings of
Nav1.5 channels (79). Recently, a new term, “late sodium
current” (INaLate), has been used to refer to this current,
which, although small in amplitude (<0.5% of the peak
INa) (79, 80), nonetheless represents an important sus-
tained depolarizing current during phase 2, thus playing
a role in maintaining the relatively long plateau of the
cardiac action potential (FIGURE 4) (78). This INaLate is
more sensitive to tetrodotoxin (TTX) and other sodium
channel inhibitors (81) than the peak INa (82).
As a steady-state component of INa, a so-called “win-
dow sodium current” in the voltage range from –65 to –
15 mV based on a different mechanism than the slow
inactivation was also suggested earlier by Attwell et al.
(9). This window current was considered to be caused
by the overlap between the steady-state activation and
inactivation curves. At present, it is still not clear whether
such a window current exists, or if the measured overlap
is due to an ultraslow inactivation or ultraslow recovery
from inactivation when this window current is deter-
mined. To better understand the nature of the sodium
current during the plateau phase of the action potential,
the possible involvement of sodium channels other than
the Nav1.5 channel, which is considered to be the major
FIGURE 4. Action potential and underlying ionic currents recorded
from human ventricular myocytes with the patch-clamp technique
cardiac sodium channel, was also suggested in several
applying human ventricular action potential as command pulses at 1 studies (83–88). As an example, Nav2.1 a-, b1-, and b2-
Hz stimulation frequency, in the absence of any sympathetic effects. subunits are highly expressed in human atrial and ven-
Inward rectifier potassium current (IK1), rapid (IKr) and slow (IKs) compo- tricular cells and Purkinje fibers (84, 85) even if their
nents of delayed rectifier potassium current, transient outward current
(Ito), and L-type calcium current (ICa,L) were measured as difference cur-
function has not been explored yet. The expression of
rent following application of selective channel inhibitors. INaL, late so- various neuronal sodium channel subtypes, e.g., Nav1.1,
dium current. Unpublished data from our laboratory at the Department Nav1.2, Nav1.3, Nav1.4, Nav1.6, and Nav1.8 (encoded by
of Pharmacology and Pharmacotherapy, University of Szeged. SCN1A, SCN2A, SCN3A, SCN4A, and SCN10A genes,
respectively), has been described in different cardiac
for the ion selectivity of the channel. The region that preparations (89), but again their functional roles are not
links domains III and IV contains the inactivation gate fully understood (3, 5, 85, 86, 90). Mishra et al. (86)
that “plugs” the channel pore after prolonged activation reported that in failing dog and human hearts neuronal
(FIGURE 3). A detailed, comprehensive review on the Nav1.1 channels were upregulated and provided signifi-
cardiac sodium channel structure has been published cant INaLate, whereas another recent study (91) showed
recently (71). Nav1.5 channels open, within a fraction of a significant upregulation of Nav1.8 channels in failing
millisecond, at potentials more positive than 60 mV, human hearts. In addition, selective inhibition of this
with strong voltage dependence. Since channel density Nav1.8 current by A-803467 (92) abolished arrhythmo-
is high, they carry a large inward current, with an ampli- genic Ca21 sparks that were attributed to enhanced in-
tude of >100 pA/pF. They also inactivate very rapidly at tracellular Ca21 load due to increased INaLate. Mutations
voltages more positive than 80 mV (at 10 mV with in the Nav1.8 encoding gene, SCN10A, were reported in
time constant s1 of 0.6 ms and s2 of 4 ms) (72), with a patients with atrial fibrillation (AF) (88) and also predis-
half-inactivation between 60 and 70 mV. Nav1.5 posed to sudden cardiac death (83). Therefore, it is

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1089


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

FIGURE 5. Regional and subcellular distribution of SCN5A/voltage-gated sodium channel (Nav)1.5 in the heart and cardiomyocytes. Left: the expres-
sion levels of SCN5A in different regions of the heart. Expression of SCN5A is highest in the atrioventricular (AV) bundle, His bundle, and right (RBB)
and left (LBB) bundle branch (dark green). SCN5A is broadly expressed in right (RA) and left (LA) atria and right (RV) and left (LV) ventricle with an epi/
endo gradient in the ventricles. SCN5A is absent from the central sinoatrial node (SAN) and atrioventricular node (AVN). Right: the localization of Nav1.5
with specific regional partner proteins in the microdomains of a cardiomyocyte: intercalated disk (ID), lateral membrane (LM), and T tubules. The sodium
current at the ID is larger than the sodium current at the LM. Reproduced from Ref. 94 with permission.

clear that further studies are needed to elucidate the accessory proteins (99). Calmodulin (CaM), the ubiqui-
role of neuronal Na1 channels in normal and diseased tous Ca21-sensing protein, plays a central role in intra-
heart (93). cellular Ca21 concentration ([Ca21]i)-dependent INa
It is known that Nav1.5 channel expression varies in function alterations by modulating fast inactivation of INa
the different regions of the heart, showing a high level of (100). Recent data suggest that CaM facilitates the re-
expression in the specialized conduction system, atria covery of the sodium channel from inactivation by inter-
and ventricles, while being absent in the central sinus acting with its inactivation gate in a Ca21-dependent
and atrioventricular nodal tissues (FIGURE 5). Also, fashion (101). The different binding sites of CaM on the
Nav1.5 channels gather in clusters and associate with sodium channel are important to understand the mecha-
accessory subunits and partner proteins, forming nisms linked to disease-associated sodium channel
region-specific macromolecular complexes (95). They mutations (102–104). In addition to CaM, Ca21/calmodu-
are not evenly distributed within the myocyte (96). The lin-dependent kinase II (CaMKII) has been shown to
current densities are large in the intercalated disk area modify INa function in a Ca21-dependent manner.
and smaller in the lateral membrane (96, 97). The com- CaMKII phosphorylation regulates cardiac Na1 channels
plex nature and the observed regional differences in so- by slowing their recovery from inactivation (105, 106).
dium channel expression, as well as the functional This results in reduced availability of fast INa at a high
significance of Nav1.5 interactions with partner proteins, rate with enhanced late INa (106). The latter contributes
justify further studies to better understand the patho- to prolonged repolarization and enhanced arrhythmia
physiology of diseases associated with Nav1.5 dysfunc- susceptibility (106, 107) often seen in heart failure, where
tion, including inherited sodium channelopathies (94, CaMKII activity is enhanced (107).
98). In addition to TTX, INa is blocked, although not sel-
The function of INa is regulated by intracellular calcium ectively, by a wide range of antiarrhythmic drugs, e.g.,
homeostasis in a complex way that involves multiple lidocaine, mexiletine, quinidine, disopyramide, and

1090 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

FIGURE 6. Tissue-specific (human) cardiac atrial, Purkinje fiber, and ventricular action potentials and the underlying ionic currents in different action poten-
tial phases, indicating their pharmacology and modulation. Black arrows indicate inward and yellow arrows indicate outward current. The contributions of dif-
21
ferent currents to the action potentials are indicated below, with a time course adjusted to the action potential. CaM, calmodulin; CaMKII, Ca -calmodulin
kinase II; hERG, human ether-à-go-go-related gene; IK1, inward rectifier potassium current; IK,Ach, acetylcholine-activated potassium current; INa, sodium current;
ICaL, L-type calcium current; ICaT, T-type calcium current; If, funny/pacemaker current; Ito, transient outward current; IKCa, calcium-activated potassium current;
IKr, IKs, and IKur, rapid, slow, and ultrarapid components of delayed rectifier potassium current; Kir, inward rectifier potassium channel; KV, voltage-gated potas-
sium channel; NaV, voltage-gated sodium channel; TASK, Tandem of pore domains in a weak inward rectifying potassium channel (TWIK)-related acid-sensi-
tive potassium channel; TTX, tetrodotoxin.

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1091


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

flecainide (FIGURE 6) (108, 109) in a frequency-depend- component (sfast 50–100 ms), the Ito recovery from
ent manner. It has recently been reported that certain inactivation also has a component with a slow recovery
drugs like ranolazine and GS967 selectively inhibit time constant (on the order of seconds) that is character-
INaLate (110, 111). Therefore, these compounds would be istic of the Kv1.4 channels (137). It is interesting that in
particularly effective for long QT (LQT) syndromes, heart rabbit ventricle the Ito current is primarily conducted by
failure (HF), or hypertrophic cardiomyopathy (HCM) (110). Kv1.4 channels (138). However, the functional role of this
On the other hand, late INa can be pharmacologically channel in rabbits is still unclear, as it is inactivated most
augmented by veratrine (112), veratridine, and ATX (113). of the time at their physiological heart rate, which is
Genetic mutations that alter INa function can lead to quite high. In humans, both Kv4.3 and Kv1.4 channels can
severe, potentially lethal, conditions and have been have functional roles, particularly in the frequency-de-
shown to play a significant role in a wide array of inher- pendent modulation of the APD during both different
ited channelopathies (for recent reviews see Refs. 94, constant or following abrupt changes in cycle lengths
114–117). As an example, loss-of-function SCN5A muta- during electrical restitution (i.e., following extrasystoles
tions lead to a reduced INa peak, thus slowing impulse with different coupling intervals). In addition, there are
conduction and possibly causing conduction block. mRNA expression studies (85, 139) supporting the possi-
These mutations have been identified in 20% of bility that the Kv1.7 channels also have a functional role
patients with Brugada syndrome (BrS) (118) as well as in for Ito in human atria and ventricles. However, because
patients with sick sinus syndrome and progressive car- of the relative paucity of data regarding Kv1.7 channels
diac conduction defect (119–121). On the other hand, in the heart, further research is needed to properly eluci-
gain-of-function SCN5A mutations have been shown to date these issues.
play key roles in congenital LQT3 syndrome (122). In Human and dog Ito start activating at membrane
some cases of familial AF, both loss-of-function and potentials more positive than 30 mV, rapidly reaching
gain-of-function SCN5A mutations have been identified peak values (within 1–2 ms) and then inactivating with a
(123, 124). double-exponential time course (sfast 5 ms, sslow 25
The late sodium or window current, irrespective of ms) (140). The Kv4.3 channel-mediated Ito, unlike Kv1.4,
its basic mechanism or molecular background, is particularly recovers rapidly from inactivation in the membrane volt-
important in arrhythmogenesis (125). As an example, in age range of 60 to 80 mV, with a time constant of
HF and in HCM, INaLate is augmented (126, 127). Since 50 ms (137). In dog and human Purkinje fibers, Kv3.4
this current is an important contributor to the action channels were also described to contribute to Ito, with
potential plateau phase, its enhancement prolongs different kinetic properties compared with Kv4.3 chan-
repolarization and increases repolarization heterogene- nels (141). This raises the possibility for fine-tuning of fre-
ity (110, 128). In addition, by increasing intracellular so- quency-dependent modulation of repolarization
dium concentration, it also increases intracellular dispersion between Purkinje fibers and ventricular mus-
calcium concentration, via the Na1/Ca21 exchanger cle (142).
(NCX). This can evoke arrhythmogenic triggered activity The transient outward potassium current and the
such as early and delayed afterdepolarizations (EADs fast inactivation of INa are important contributors to the
and DADs, respectively) (129–131), discussed below in early/fast repolarization of the action potential (phase
this review. Both triggered automaticity and enhanced 1) (137, 143) (FIGURE 4). Phase 1 repolarization and Ito
dispersion of repolarization are considered major mech- are more prominent in Purkinje fibers and atrial, mid-
anisms in arrhythmogenesis, and their reduction is a myocardial, and subepicardial ventricular muscle (137),
major aim in antiarrhythmic drug development. but they are small, or nonexisting, in subendocardial
ventricular cells and sinoatrial (SA) and atrioventricular
3.2. The Transient Outward Current (AV) nodal cells (137). Interestingly, phase 1 repolariza-
tion and Ito are not present in guinea pig ventricular
The main channels providing the transient outward po- myocytes (144, 145). Also, it has been reported that in
tassium current (Ito) in human and dog ventricular muscle mouse ventricle Ito is small and action potential is long
are Kv4.3 and Kv4.2 pore-forming a-subunits coas- after birth and later the action potential shortens as Ito
sembled with KChIP2 and DPP auxiliary subunits (85) develops in adult mice (146, 147). The impact of Ito on
encoded by KCND3, KCND2, and KCNIP2 and DPP6/10 the shape of the action potential waveform is complex.
genes, respectively (132–134). Kv1.4 a channel subunits In addition to its role in phase 1, Ito also modulates the
(encoded by KCNA4) are also expressed with marked voltage level of the plateau, and consequently it has
regional and interspecies dependence, making up 10– an indirect influence on activation, inactivation, and
20% of Ito density in humans (135, 136). Accordingly, in deactivation of several other transmembrane ionic cur-
humans (unlike in dogs), in addition to the rapid rents that operate during the plateau phase. Since Ito,

1092 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

similarly to INa, has activation, deactivation, and inacti- Ito an important contributor to repolarization abnormal-
vation properties, it was also suggested that a window ities in altered thyroid status.
Ito current may be generated, in the membrane poten-
tial range from 35 to 10 mV. This window current 3.3. Inward Calcium Current
would also contribute to the final repolarization (140). It
was also reported that interaction of DPP10a with The inward calcium current (ICa) in the heart was first
Kv4.3 channels results in a sustained current compo- described by Reuter (170), and it has two major types
nent of human atrial Ito that can participate in the late (171, 172). The most abundant type is the L-type calcium
repolarization phase (148). However, despite its poten- channel (Cav1.2), which conducts current through the
tial importance, it is difficult to validate or disprove the pore-forming a-subunit (a1) encoded by the CACNA1C
existence of such current, since selective inhibitors are gene (85). The a1-subunit consists of 2,000 amino
lacking. This suggests the need for further research to acids, organized into four repeat domains (I–IV), each
clarify this issue. Ito is a typical transmembrane ionic containing six transmembrane segments (S1–S6) (173).
current that can be attributed to the function of a wide The S4 transmembrane helix from each domain collec-
variety of distinctly different potassium and also chlo- tively constitute the voltage sensor of the channel (173).
ride ion channels (137). Although the role of Ito in A region called the “P loop” connecting the S5 and S6
arrhythmias is not well defined, it definitely plays a role segments is responsible for the Ca21 selectivity of the
in Brugada syndrome (117, 149), where a reduced pore region (174). The pore-forming subunit coassem-
expression level of the auxiliary Ito subunit, KChIP2, in bles with the extracellular a2d and intracellular b auxil-
females most likely underlies the male phenotypic pre- iary (predominantly b2 in cardiac tissue) subunits that
dominance (150, 151). The stronger epicardial Ito in modulate kinetics, gating, and trafficking properties of
males could further aggravate the consequence of the channel (175–177). Inward L-type Ca21 current (ICa,L)
impaired INa in Brugada patients; in this case, inhibition has a very rapid activation (14) and is particularly impor-
of Ito would be beneficial. Multiple nonselective Ito tant for excitation-contraction coupling (178), since it
inhibitors have been reported (FIGURE 6): 4-aminopyr- serves as a trigger for the calcium-induced calcium
idine (4-AP), in millimolar concentrations (152); quini- release (CICR) (179) from the SR and as a source of
dine, flecainide, and chromanol 293B, in micromolar extracellular calcium when needed. In addition, ICa,L
concentrations (140, 152); and phrixotoxin, in nanomo- plays a fundamental electrophysiological role in main-
lar concentrations (153). However, a selective Ito inhibi- taining the plateau phase of the action potential
tor is still lacking. This could constitute a potential (FIGURE 4) and in the depolarization of SA and AV nodal
treatment in atrial fibrillation, by increasing APD and cells (180). In these cell types, ICa,L is the main contributor
consequently ERP, in atrial but not or less in ventricular to impulse conduction; therefore its impairment pro-
tissue (154). An activator for Ito was also reported hav- longs the PQ interval and can result in AV node conduc-
ing effect on canine ventricular but not atrial myocytes tion block. In addition, since ICa,L provides depolarizing
(155). current, its decrease would reduce the spontaneous fre-
Ito is downregulated in HF (156–159), HCM (160), and quency of these cells. In AV nodal cells, this shift in
diabetes mellitus (161, 162), possibly contributing to threshold potential also contributes to the slowing of AV
repolarization prolongation in these pathological set- impulse conduction. The inactivation kinetics of L-type
tings. Recently, it was shown that Kv4.3 (fast Ito) and ICa (sfast 2–8 ms, sslow 30–100 ms) depend not only
Kv1.4 (slow Ito) were expressed differently in normal on membrane voltage but also on the intracellular Ca21
and failing hearts, thus contributing to arrhythmogenic concentration (14, 15, 181–184), which is dynamically
regional heterogeneity in action potential waveforms changing during the action potential. The recovery from
(101). It was also demonstrated that a slowing of phase inactivation is complex and strongly depends on volt-
1 repolarization, which can be due to the decreased Ito age. At 40 mV it can be characterized by an exponen-
often observed in failing hearts (163–165), decreases tial time course ranging between 30 and 60 ms with
the driving force of Ca21 through L-type calcium chan- sfast = sslow (14, 185). Importantly, the recovery kinetics
nels, and it can result in potentially arrhythmogenic can be faster in ventricular and Purkinje fibers, which
asynchronous Ca21 release from the sarcoplasmic have a resting potential around 80 mV. The ICa,L was
reticulum (SR) (166). Ito is subject to a- and b-adrener- originally called slow inward current (Isi), since with the
gic regulation, both decreasing Ito via the PKA and old experimental methods, before the introduction of
PKC pathways (167), whereas CaMKII has been shown single-cell voltage clamp, a relatively slow ICa activation
to increase Ito (167). was measured (170, 186). Later, with more advanced
Thyroid-stimulating hormone and thyroid hormones voltage-clamp techniques, the proper fast activation
have been shown to modulate Ito (168, 169), thus making kinetics could be determined (187).

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1093


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

Table 1. Changes of genes, channel/transporter proteins, and ionic currents in various genetic disorders
Genetic Disorder Gene Protein Ionic Current/Function

Trigger

CPVT 1 RYR2 Ryanodine receptor SR Ca21 release

CPVT2 CASQ2 Calsequestrin : SR Ca21 release

Inherited sinus bradycardia SCN5A Nav1.5 ; INa

Sick sinus syndrome HCN4/SCN5A HCN4/Nav1.5 ; If/INa

Familial inappropriate tachycardia HCN4/SCN5A HCN4/Nav1.5 : If/INa

Substrate

Brugada syndrome SCN5A Nav1.5 ; Impulse conduction

ARVC (Naxos disease) Desmosome protein ; Impulse conduction

LQT1 KCNQ1 KCNQ1 (Kv7.1) ; IKs

LQT2 KCNH2 hERG (Kv11.1) ; IKr

LQT3 SCN5A Nav1.5 : INa

LQT4 (ankyrin-B syndrome) ANK2 Ankyrin-B Multichannel interactions

LQT5 KCNE1 KCNE1 (minK) ; IKs

LQT6 KCNE2 KCNE2 (MiRP1) ; IKr

LQT7 (Andersen–Tawil syndrome type 1) KCNJ2 Kir2.1 ; IK1

LQT8 (Timothy syndrome) CACNA1C Cav 1.2 : ICa

LQT9 CAV3 Caveolin 3 : INa

LQT10 SCN4B Nav 1.5 b4 : INa

LQT11 AKAP9 AKAP-9 (yotiao) ; IKs

LQT12 SNTA1 a1-Syntrophin : INa

LQT13 KCNJ5 Kir3.4 (GIRK4) ; IKACh

LQT14 CALM1 Calmodulin Multichannel interactions

LQT15 CALM2 Calmodulin Multichannel interactions

LQT16 CALM3 Calmodulin Multichannel interactions

SQT1 KCNH2 HERG : IKr

SQT2 KCNQ1 KVQT1 : IKs

SQT3 KCNJ2 Kir2.1 : IK1

SQT4 CACNA1C Cav a1 ; ICa,L

Continued

1094 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

Table 1.—Continued
Genetic Disorder Gene Protein Ionic Current/Function

SQT5 CACNB2b Cav b2b ; ICa,L

SQT6 CACNA2D1 Cav a(2) d-1 ; ICa,L

SQT7 SLC22A5 OCTN2 Carnitine deficiency

SQT8 SLC4A3 AE3 ; Cl /HCO3 exchanger function

Familial AF KCNQ1 KVLQT1 ; IKs

KCNE2 MIRP1 ;?

KCNJ8 Kir6.1 : IK,ATP

AF, atrial fibrillation; ARVC, arrhythmogenic right ventricular cardiomyopathy; CPVT, catecholaminergic polymorphic ventricular tachycardia; ICa, calcium
current; ICa,L, L-type Ca21 current; If, funny/pacemaker current; IKACh, acetylcholine-activated potassium current; IK,ATP, ATP-sensitive potassium current;
IKr, rapid component of delayed rectifier potassium current; IKs, slow component of delayed rectifier potassium current; IK1, inward rectifier potassium cur-
rent; INa, sodium current; LQT, long QT syndrome; SQT, short QT syndrome; SR, sarcoplasmic reticulum.

ICa,L is modulated (FIGURE 6) by cAMP-dependent fibers is still unclear. However, it plays an important role
phosphorylation and other factors, including intracellular in the SA and AV nodal cells (211), where it makes a sig-
Ca21 levels (15). It has been shown that CaM supports nificant contribution to the pacemaker function. This cur-
both inactivation and facilitation of ICa (188). The intracel- rent is conducted by Cav3.1 and Cav3.2 channels,
lular Ca21 enhances ICa via CaMKII, involving direct encoded by CACNA1G and CACNA1H genes, respec-
phosphorylation of L-type Ca21 channels (189, 190) inde- tively (212). ICa,T activates at more negative membrane
pendently of cAMP via PKA (191) involving Rad, a mono- potentials than ICa,L, and its overlap with INa makes it diffi-
meric G protein that closely interacts with Cav1.2 (192). cult to study (172). ICa,T can be inhibited by low concen-
ICa,L can be effectively blocked (FIGURE 6) by Cd21, ve- trations (100–200 mM) of Ni21 (172) and by the organic
rapamil, and diltiazem, but the inhibition with these compound mibefradil, which was developed with the
drugs is not selective (193–195). Dihydropiridines, e.g., aim of decreasing elevated heart rate (213).
nifedipine (196) and nisoldipine (197), are more selective
ICa,L blockers, but they may be sensitive to light (198). 3.4. Delayed Rectifier Potassium Currents
Pharmacological activation of ICa,L is also possible by
Bay K8644 (FIGURE 6) (199). Before the introduction of the patch-clamp technique, a
The ICa,L has key roles in several diseases like HF slowly activating current carried by K1 was recorded
(200), HCM (27), AF (201), and myocardial ischemia and during the plateau phase. This current was named the
in other pathophysiological conditions, such as the de- delayed rectifier potassium current (Ix1/2). Later, by apply-
velopment of EADs, DADs, (202–204) and cardiac is- ing single-cell patch-clamp technique, Sanguinetti and
chemia-reperfusion (205), discussed in more detail in Jurkiewicz (214) showed that this delayed rectifier out-
other parts of this review. ward potassium current can be separated into a rapid
A gain-of-function G406R mutation of the Cav1.2 chan- (IKr) and a slow (IKs) component. Molecular biological
nel causes type 8 of congenital LQT syndrome, also studies also confirmed that these two IK components are
called Timothy syndrome (TABLE 1). This disease is conducted by distinctly different ion channels.
characterized by slower inactivation of ICa,L (206–208)
and by the fact that small clusters of Cav1.2 channels
have a larger probability for coordinated opening and 3.4.1. The rapid delayed-rectifying potassium
closing (“coupled gating”) (209), thus leading to tachyar- current.
rhythmia and congenital heart defects (ductus arterio-
sus, ventricular septal defect, Fallot tetralogy, HCM) The rapid delayed rectifier outward potassium current
(210). (IKr) is conducted by the Kv11.1 pore-forming a-subunit,
The second type is the T-type calcium current (ICa,T), also called hERG in humans (human ether-à-go-go
for which significantly less data are available. Its func- related gene), which is associated with various acces-
tional role in atrial and ventricular cells and Purkinje sory b and possibly other subunits (85, 215, 216).

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1095


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

Similarly to other Kv channel pore-forming a-subunits, other b-subunits like MinK, MiRP2, MiRP3, and MiRP4
Kv11.1 consists of six transmembrane segments (S1–S6) were abundantly expressed in human atrial and ven-
and the functional channel contains four a-subunits tricular tissue and Purkinje fibers (85). Coexpression of
(FIGURE 3) (217). The Kv11.1 hERG a-subunit has two iso- the b- and hERG a-subunits produced currents with
forms (a and b), which are different in terms of gating kinetics similar to those native currents that can be
and drug sensitivity (65, 66). The wide variety of interact- recorded in different species including human (219,
ing accessory b-subunits include MinK (human minimal 220). However, the exact role of these b and other
potassium ion channel), MiRP1 (mink-related peptide 1), possible accessory subunits, how they regulate IKr,
and MiRP2, MiRP3, and MiRP4 proteins encoded by and whether they are responsible for the observed
KCNE1, KCNE2, KCNE3, KCNE4, and KCNE5 genes, species-dependent differences and drug sensitivities
respectively (85, 215). Initially, it was suggested that are unclear at present and need to be elucidated in
MiRP1 interacted with hERG (Kv10.1) to form IKr channels the future. IKr activates in a voltage-dependent man-
(218). This is due to the fact that when hERG (Kv11.1) a ner, with an activation s of 31 ms at 130 mV in human
channel subunits were expressed alone in HEK cells, a ventricular myocyte (221). It also slowly deactivates
very rapidly activating steady-state-like current was (222) in a voltage-dependent manner; its deactivation
observed, and only coexpression with MiRP1 resulted in can be fitted by a double exponential with sfast of 600
currents that resembled native IKr. On the basis of this ms and sslow of 6,800 ms at 40 mV. The ratio of the
observation MiRP1 was considered the most important amplitudes of the fast and slow components increases
accessory subunit to form the native IKr channel. at more negative potentials. IKr exhibits a peculiar, very
However, later studies indicated very low levels of rapid, inactivation (223–225), which starts even before
KCNE2 expression in human heart, whereas genes of it activates.

FIGURE 7. The role of the slow component of the delayed


rectifier potassium current (I ) in the repolarization in dog
Ks
ventricular papillary muscle (A–C) and in dog single ventricu-
lar myocyte (D). In A, action potential duration (APD) is not,
or minimally, changed after full I inhibition by 100 nM L-
Ks
735,821 without external sympathetic stimulation. In B, in the
same preparation full IKs inhibition elicited significant prolon-
gation of repolarization in a preparation where the rapid
component of the delayed rectifier potassium current (IKr)
was inhibited by E-4031 and late sodium current (I ) was
NaLate
augmented by veratrine. In C, transmembrane current
recordings show that during a short (150 ms) voltage pulse
very little I develops, but when pulse duration was
Ks
increased to 500 ms significant IKs developed, explaining
the lack and significant changes of APD after I inhibition in
Ks
A and B. (Reproduced from Ref. 228 with permission.) In D,
the result of a representative experiment is shown in dog
ventricular myocytes. In the baseline (control) situation, small
short-term APD variability and normal APD were recorded
that did not change significantly after I inhibition by chro-
Ks
manol 293B. Additional application of I block by almoka-
Kr
lant increased both APD and short-term APD variability
illustrated by the Poincare plot, which was substantially fur-
ther lengthened and increased by additional IKs inhibition,
respectively. (Reproduced from Ref. 255 with permission.)

1096 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

During the action potential in early plateau and in of IKr-blocking drugs (236). The effect of ischemia on IKr
phase 3, IKr channels rapidly recover from inactivation is complex, since acidosis was reported to decrease IKr
and they reopen as voltage changes toward more nega- (237, 238), particularly by inhibition of the hERG1b iso-
tive values, and deactivation progresses. Accordingly, de- form of the channel (239), but hyperkalemia can
spite the rapid activation of the current during the plateau increase (234, 235) the current—and both are present in
phase, a relatively tiny current develops that gradually ischemia. Reduced IKr was also reported in the infarcted
increases and decreases during phase 3 repolarization zone in dog myocytes (240). In HF, IKr is generally con-
(FIGURE 4); therefore it is a crucially important current to sidered downregulated (241), even if different studies
secure repolarization. report sometimes contradictory results.
Since IKr deactivates slowly, it does not have time to Loss-of-function mutations in hERG channel can cause
fully deactivate during an action potential. Therefore, a congenital long QT syndromes (237, 242, 243), whereas
residual and gradually decreasing outward current can gain-of-function mutations lead to short QT syndromes
still flow through this channel, thus shortening the next (244–246) (TABLE 1).
action potential when diastolic interval is relatively short,
i.e., during fast heart rate or during an early extrasystole 3.4.2. The slow delayed-rectifying potassium
(222). Consequently, IKr is considered a key player in fre- current.
quency-dependent APD regulation, and it can influence
the pacemaker function as well (226). The slow component (IKs) of IK is carried by the Kv7.1 chan-
IKr can be blocked by specific compounds in the sub- nel, consisting of a pore-forming a-subunit (FIGURE 3)
micromolar or micromolar range (e.g., dofetilide and E- (217), encoded by the KCNQ1 gene, that coassembles
4031) (FIGURE 13 and FIGURE 7), causing a marked pro- with various MinK, MiRP1, MiRP2, MiRP3, MiRP4, and
longation of the action potential (227, 228). It was other accessory subunits, encoded by KCNE1, KCNE2,
reported that this current can be modulated (FIGURE 6) KCNE3, KCNE4, and KCNE5 genes, respectively (85,
by endogenous substances like endothelin, which sup- 215). MinK was the first b accessory protein (247, 248)
presses IKr (229). Decreased IKr was also observed after that was identified for the Kv7.1 channel, but later the im-
a1- and b1-receptor activation, linked to the PKC and PKA portance of other b-subunits (e.g., MiRP1, MiRP2, MiRP3,
pathways, respectively (230–232). There are also some MiRP4) was recognized, suggesting that this variability in
compounds known to enhance IKr (FIGURE 6). b-subunits may serve as the basis for the marked inter-
In pathophysiological conditions, IKr can change. In hy- species variability in native IKs properties (67, 68). In
pokalemia, the magnitude of the current decreases guinea pig, the amplitude of IKs is large [(3, 214, 249), and
(233–235), thus making the heart vulnerable to torsades its kinetics differs from those measured in rabbit (250,
de pointes (TdP) arrhythmia, especially in the presence 251), dog (228) or human (252, 253) ventricular muscle. In

FIGURE 8. A–C: influence of slow compo-


nent of delayed rectifier potassium current
(IKs) inhibition by chromanol 293B (A), sym-
pathetic stimulation by isoproterenol (ISO,
B), and combination of IKs block and sympa-
thetic stimulation (C) in isolated dog cardiac
Purkinje cells. Note that in control (Ctl) con-
dition plateau phases of the Purkinje cells
are at more negative voltage than the acti-
vation threshold of IKs, and accordingly IKs
inhibition did not change repolarization. In B,
isoproterenol shifted the action potential
plateau voltage to more positive values
because of substantially increased IKs. D: in
this situation, IKs inhibition substantially len-
gthened repolarization as shown in C. (Rep-
roduced from Ref. 157 with permission.)

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1097


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

most species, IKs activates slowly and in a voltage-de- (255, 265). PKC has a biphasic influence, increasing IKs after
pendent manner, at potentials more positive than 30 short-term application and decreasing IKs after long-term
mV, with s of several hundred milliseconds to seconds application (266). It was also reported that the PKC-medi-
(254). The current deactivates rapidly, except in guinea ated IKs augmentation was PKC isoform dependent (267,
pigs, in a voltage-dependent manner, with s of <200 ms 268). Drugs are available to inhibit (228) and to activate
at 40 mV (253). The density of IKs during the plateau of (269) IKs (FIGURE 6). Hypokalemia and elevated intracellular
a normal action potential is small (FIGURE 4), because of Ca21 enhance and hyperkalemia decreases IKs (235, 270).
its slow activation. Therefore, its contribution to repolari- Diseases like heart failure (7) and diabetes mellitus
zation without b-adrenergic stimulation is minimal, except (271) and genetic mutations (LQT1) (114, 149) or drugs
in guinea pig (FIGURE 7A). However, normal physiologi- (228) can reduce IKs channel expression, and, as a con-
cal conditions always involve some level of sympathetic sequence, the reserve repolarizing current provided by
tone that increases basal IKs density (255). In addition, IKs would be diminished.
when plateau voltage is enhanced by the elevated sym- This scenario can be further aggravated when intracel-
pathetic tone, more IKs develops and contributes more to lular cAMP is elevated, and cAMP-dependent phospho-
repolarization (256) (FIGURE 8). IKs is a critical contributor rylation enhances L-type ICa. Since ICa,L is an inward
to the repolarization reserve (257, 258). According to this current, which prolongs repolarization, the negative feed-
concept, cardiac repolarization is provided by multiple back function of impaired IKs cannot sufficiently limit ex-
redundant ionic currents that can compensate for each cessive APD prolongation. Therefore, dispersion of
other’s loss of function. Therefore, a dysfunction of a sin- repolarization and propensity for life-threatening arrhyth-
gle repolarizing current does not necessarily cause a mia can increase (272). Genetic mutations can also lead
measurable effect on repolarization. However, congenital to gain of function of IKs, resulting in increased current.
or acquired defects in the function of multiple currents The mutations have been shown to have concomitant
can be additive. As shown in FIGURE 7, A and D, IKs block effects in sinoatrial, ventricular, and atrial cardiomyocytes
at baseline does not prolong ventricular repolarization and lead to complex clinical phenotypes (273–281).
and does not increase short-term variability of APD, which
is a recently suggested predictive parameter of proar- 3.5. Inward Rectifier Potassium Current
rhythmic risk (259). However, after IKr block (by E-4031 or
almokalant), IKs inhibition significantly prolongs repolariza- Inward rectifier potassium current (IK1), similarly to Ito, is a
tion (FIGURE 7B) and increases short-term variability of current that is carried by several channels (282), even if
repolarization. At longer repolarization duration, induced this fact is sometimes overlooked when its behavior is
either by IKr block or by longer voltage pulses (FIGURE 7, discussed (283). Therefore, the definition of IK1 needs
B and C), more IKs develops and, as a negative feedback
mechanism, it can limit excessive APD lengthening (221,
228, 260). Although some studies have reported IKs in I (pA)
atrial tissue (261, 262), and both KCNQ1 and its accessory 800

subunits are expressed in the atria at levels similar to


those in the ventricle (263), the presence and role of IKs in
400
human and canine atrial including sinoatrial node (SAN)
cells are still unclear. Assuming similar IKs kinetics in atria
and ventricles, less IKs activation can be expected in atrial V (mV)
myocytes, since the plateau phase is shorter and it devel- -120 -100 -80 -60 -40 -20 20
ops at more negative voltages than in ventricular myo- Kir2.3 Untransfected cell
cytes. One may speculate that at very rapid rates (e.g., Kir2.1 -400
during atrial fibrillation or flutter) the continuous presence
of a very tiny IKs current during the action potential, due to
frequent channel opening and slow recovery compared -800

with the diastolic interval, may produce a steady-state- Kir2.2


like sustained outward current, which may have a role in
-1200
modulating the resting membrane potential, the pace-
maker activity, or even the APD. To resolve these contro- FIGURE 9. Distinct current (I)-voltage (V) relations of inward recti-
versies, further research is needed. fier potassium (Kir)2.x channels expressed in HEK293 cells. Different
colors depict ramp ( 100 to 0 mV)-generated, barium-sensitive cur-
IKs is regulated by endogenous substances (FIGURE 6).
rents in cells expressing Kir2.x channels. The current in an untrans-
Progesterone, and cAMP/PKA-dependent phosphorylation fected cell is shown in green. (Reproduced from Ref. 63 with
via the Yotiao accessory subunit protein (264), increases IKs permission.)

1098 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

particular care. Here we define IK1 as a current con- by the organic compound PA-6 in the nanomolar range
ducted by channels consisting of Kir2.1, Kir2.2, Kir2.3, and (298).
Kir2.4 pore-forming a-subunits, encoded by KCNJ2, The autosomal dominant mutation of KCNJ2 causes
KCNJ12, KCNJ4, and KCNJ14 genes, respectively (215, Andersen–Tawil syndrome (LQT7), having a characteris-
282). Four a-subunits (each containing 2 transmem- tic triad of episodic flaccid muscle weakness, ventricular
brane segments, S1 and S2) combine to form the tetra- arrhythmias, and prolonged QT interval (299) (TABLE 1).
meric, functional Kir channel (FIGURE 3) (217). IK1 Pharmacological approaches to investigate KCNJ2 loss-
channels may also have other, as yet undefined, acces- of-function mutation revealed that the IK1 blocker BaCl2
sory subunits. Remarkable differences have been prolonged the QT interval without causing a major
observed between currents flowing through the differ- increase in transmural depolarization. EADs and sponta-
ent Kir channel isoforms (63) expressed in mammalian neous torsades de pointes arrhythmias were not
cell lines (FIGURE 9), thus raising the possibility that dif- observed, nor could they be induced by electrical stimu-
ferent species and cardiac tissue types express different lation. These observations may explain why QT prolon-
channel isoforms, with various phenotypes. Further gation of the Andersen Tawil syndrome was found to
research will elucidate the exact roles of different Kir be relatively benign in clinical settings (300).
channel isoforms. The upregulation of Kir2.1 was observed in atrial fibril-
IK1 is considered one of the most important currents lation, where it may contribute to APD shortening (205).
securing the resting membrane potential and shaping In contrast, Kir2.1 downregulation was reported in HF,
terminal repolarization in phase 3 of the action potential leading to APD prolongation and facilitating delayed
(FIGURE 4). According to classical electrophysiological afterdepolarization (DAD) formation (157). Sections 7.1,
principles, one would expect potassium channels to 7.2, 7.3, and 7.5 provide more detailed information
conduct current more easily in the outward direction, as regarding the relevance of altered IK1 function and
the intracellular K1 concentration is much greater than expression in several diseases.
the extracellular. Instead, inward rectification means that
the steady-state current decreases at potentials positive 3.6. Ultrarapid Delayed Rectifier Current
to about 50 mV. This inward rectification was attrib-
uted to channel inactivation caused by intracellular The channel carrying the ultrarapid delayed-rectifying
Mg21 and polyamines (284, 285). At potentials more potassium current (IKur) was first cloned from human ven-
negative than the K1 reversal potential, IK1 is an inward tricles (301) but is markedly more expressed in atria
current that has a rapidly inactivating component (286) (302). At first, it was considered to be a sustained com-
negative to 120 mV, but the physiological importance ponent of Ito (303)—“Ito,sus.” However, this current is car-
of this inactivation has not been established. This cur- ried by a distinct channel consisting of one Kv1.5
rent is regularly measured as a Ba21-sensitive steady- a-subunit, encoded by the KCNA5 gene, and Kvb1.2 and
state current at the end of voltage pulses (287). 1.3 accessory subunits (304, 305).
However, Ba21 is not fully selective for any of the K1 IKur is activated rapidly upon depolarization at mem-
channels described so far, and the steady-state current- brane potentials more positive than 0 mV, with an activa-
voltage relation measured can also include other, not tion s of 13 ms at 0 mV potential (302). IKur inactivates
well-characterized, currents, described as “leak” or “pla- slowly at depolarized potentials, with a double-exponen-
teau” currents (Ileak or Ip, respectively) (4, 283, 288). tial s (609 ms and 5,563 ms, respectively, at 140 mV
There are reports suggesting that intracellular [Ca21] voltage) (306), and its inactivation can be enhanced by
changes can modulate IK1 (287, 289, 290), possibly rele- sympathetic stimulation (307). This current is thought to
vant to frequency-dependent APD regulation or in cer- operate during phases 1–3 of the action potential of
tain pathological settings (291). IK1 is also increased by atrial cardiomyocytes and Purkinje fibers but not in ven-
elevated extracellular K1 concentration ([K1]) (292, 293). tricular muscle. However, this was challenged by the
Hyperkalemia increases IK1 by electrostatically destabi- group of Carnes (308), who measured IKur as a 4-AP-sen-
lizing Mg21- and polyamine-induced IK1 block by displac- sitive current in dog ventricular myocytes. In these dog
ing these cations from their binding sites (294). ventricular myocytes, 50 mM 4-AP also lengthened APD.
Activation of both the a1/PKC and b1/PKA pathways have These findings, however, need to be confirmed by other
been shown to reduce IK1 in human isolated cardiomyo- investigations. In connection with this, similar mRNA
cytes (295, 296). A similar effect of neurokinin-3 was expression levels were measured in ventricles and in
observed on IK1 in rabbit and human atria (297). These Purkinje fibers, but one order of magnitude smaller than
results further emphasize the importance of autonomic in atria (85). Since mRNA measurements from cardiac
control of atrial APD and electrical function. IK1 can be inhib- samples may be contaminated by coronary vessel neu-
ited (FIGURE 6) by Ba21 in the micromolar range (140) and ronal tissue and also mRNA from fibroblasts, whether

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1099


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

IKur exists and/or operates in ventricular muscle is still expression, this current was not detected, nor was APD
not fully clarified (252). This question is particularly im- lengthened by apamin, the most established inhibitor of
portant because the impact of IKur on the cardiac repola- this channel, in rat, dog, rabbit, and human undiseased
rization seems complex (309). After the discovery of IKur, ventricular muscle, thus bringing into question its role in
it was thought that selective IKur inhibitors would have a physiological conditions (321). However, other studies
particular advantage in the treatment of atrial fibrillation, have reported an important role for SK2 channels in the
by lengthening APD and ERP in the atria but not in the human atria.
ventricles. Despite promising animal experiments, these Most of the evidence regarding the highly apamin-
expectations have not been fulfilled, since clinical stud- sensitive SK2 current originates from experiments
ies did not result in convincing outcomes with newly carried out in mice, rats, or rabbits (313, 315, 322,
developed IKur blockers (FIGURE 6), e.g., XEN-D0101 323). In these species, atrial cellular electrophysiol-
and XEN-D0103 (310, 311). Inhibition of IKur by XEN-D0101 ogy is not well explored and may differ from dogs or
and XEN-D0103 (310) moderately but significantly humans. In addition, a marked chloride current was
increased APD in atrial preparations from patients with described in rabbit atria that was not found in dog or
atrial fibrillation and remodeling, whereas no significant human atria (324). Furthermore, some of the evi-
changes were observed in preparations from patients dence comes from drug studies performed with
with sinus rhythm (309). These experiments also NS8593, a putative selective inhibitor of SK2 chan-
showed that IKur inhibition shifts the atrial plateau phase nels (5), whose effects on all important ionic currents
toward the positive direction, thus potentially affecting in native cardiac cells have not been tested. The-
activation, inactivation, or deactivation of other plateau refore, further investigations are needed for a better
currents, most notably IKr. Thereby, IKur inhibition may understanding of the exact role of SK2 current in car-
produce variable effects of APD and repolarization diac physiology and pathophysiology, which is cur-
depending on tissue and experimental conditions (154). rently a very interesting and important issue.
This could have important implications if IKur is actually
present in the ventricles, where repolarization reserve 3.8. Ligand-Gated Ion Channels
would be decreased by IKur inhibition, with a consequent
increase in proarrhythmic risk. IKur is under adrenergic 3.8.1. ATP-sensitive potassium channels.
control: a1-receptor stimulation reduces and b1-receptor
stimulation increases this current in atrial myocytes (306). ATP-sensitive potassium channels, carrying the IK,ATP
current, were first identified in cardiac sarcolemmal
3.7. Calcium-Activated K1 Current membrane by Noma in 1983 (325). KATP channels were
first cloned by Aguilar-Bryan and colleagues (92) and
This current is conducted by small-conductance cal- comprise heterooctamers consisting of four inward-rec-
cium-activated potassium channels (SK2 or KCa2.1–2 tifying potassium channel pore-forming subunits (Kir6.1
and 3), which constitutively bind calmodulin and are or Kir6.2, encoded by KCNJ8 and KCNJ11 genes, respec-
encoded by KCNN1, KCNN2, and KCNN3 genes (5). The tively) and four ATP-binding cassette protein sulfonyl-
question of whether a calcium-activated K1 current urea receptors (SUR1 or SUR2, encoded by ABCC8 and
exists in cardiac tissue and what impact it could have on ABCC9 genes, respectively) (326). The SUR2 has two al-
the action potential was raised long ago (312). Although ternative RNA splice variants, SUR2A and SUR2B; the
>35 yr have passed since then, and its existence is now two polypeptides differ in their COOH terminals (327).
well established in various species and tissues (313), its Different tissues express KATP channels with distinctly
physiological and pathophysiological roles in cardiac different subunit composition, thus leading to distinct
muscle are still not fully elucidated. Most evidence sug- pharmacological properties of these channels; the myo-
gests that this current plays a role in the atria (5, 314), cardial plasma membrane KATP channel consists of
and it is also implicated in diseases such as atrial fibrilla- Kir6.2/SUR2A (327). At first glance, their nomenclature
tion and heart failure (5, 315–317). A recent study in rat might be misleading: these channels are kept closed by
ventricle showed that SK2 channels are upregulated in physiological intracellular ATP levels in normoxic cardiac
HF and also enhanced by PKA phosphorylation (318). In tissue, and they activate during metabolic stress, when
atrial myocytes isolated from patients with AF, SK2 cur- the ratio of ATP to ADP is decreased, e.g., in myocardial
rent was increased by enhanced activation of CaMKII ischemia (328). This makes these channels unique,
(319). The calcium-activated K1 current is activated in a directly connecting cellular metabolism to membrane
voltage-independent manner or in response to intracel- excitability. KATP channels are not regulated by mem-
lular Ca21 concentration increase, with a half-maximal brane voltage or calcium levels. Indeed, they belong to
activation of 300 nM [Ca21]i (320). Despite SK2 channel the inward rectifier (Kir) potassium channel group, with

1100 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

IK,ATP exhibiting weak inward rectification properties on rectifying current, the acetylcholine-activated potassium
the current-voltage relationship that is regulated by in- current (IK,ACh), and are largely expressed in atrial, SA,
tracellular Mg21 and polyamines (329–331). Activation and AV nodal cells (355, 356). IK,ACh (355, 356) is particu-
of sarcolemmal IK,ATP during myocardial ischemia heter- larly important in atrial, sinus, and AV nodal cells where
ogeneously shortens the action potential, and may pro- they are abundantly expressed. Genes for Kir3.1 and
mote reentry (31). Accordingly, several investigations Kir3.4 are also present in significant amounts in human
found KATP activation to be proarrhythmic (332), thus cardiac Purkinje fibers (85), consistent with the observa-
suggesting sarcolemmal IK,ATP inhibition for the preven- tion that acetylcholine and carbachol shorten APD via
tion of myocardial ischemia and ischemia-reperfusion- IK,ACh in atria, SA, and Purkinje fibers (357). However, its
induced arrhythmias (333–335). In contrast, some stud- influence on repolarization in the ventricles is uncertain
ies found no antiarrhythmic effects of KATP blockers in (205), and its expression level is relatively low (85). It
this setting (336, 337). Furthermore, pharmacological must be emphasized that increased parasympathetic
KATP channel activation was also found to exert antiar- tone and adenosine can also indirectly decrease ICa,L am-
rhythmic effects (200, 338, 339) that could be, at least in plitude, through the G inhibitory protein decreasing
part, be explained by reduction of monophasic action cAMP, thus contributing to shortening of repolarization
potential duration heterogeneity (340) and decreased and shifting the plateau potential toward the negative
triggered activity in Purkinje fibers upon KATP opener direction. The IK,ACh-induced repolarization shortening is
administration after myocardial infarction (341, 342). It generally considered an important factor in the develop-
should be also emphasized that KATP channel activation ment of atrial fibrillation (358), and this initiated intensive
is very important for prolongation of cell survival during efforts in the drug industry to develop specific IK,ACh inhib-
ischemia, since, according to the classical hypothesis, itors (359). Cholesterol was also reported to enhance
the consequent action potential shortening would IK,ACh (360). The IK,ACh current has an important role in
reduce contractility by reducing calcium entry into the modulating the resting or maximal diastolic potential as
cardiomyocytes and enhancing Ca21 extrusion via for- well, thus hyperpolarizing atrial, SA, and AV nodal tissues
ward-mode NCX activity (343–345). However, cardio- and Purkinje fibers. This effect interferes with the pace-
protection following sarcolemmal IK,ATP activation in maker function, slowing the rate of spontaneous automa-
quiescent myocardial cells subjected to hypoxia was ticity (361), and it could also slow conduction in the SA
also observed (346), suggesting that decreased Ca21 and AV nodes, since hyperpolarization can increase the
overload via reduced reverse-mode NCX activity during potential difference between the maximum diastolic
resting membrane hyperpolarization (347) can also con- potential and threshold of activation of ICa,L, which is re-
tribute to sarcolemmal KATP-mediated cardioprotection. sponsible for depolarization in these tissues.
Therefore, the effects of KATP channel modulation on is- It has been reported that IK,ACh is constitutively active
chemia and ischemia-reperfusion-associated cardiac (362) in atrial cells isolated from patients with chronic
arrhythmias remains controversial. atrial fibrillation (358) and therefore plays a pivotal role
In addition to the KATP channels found in the sarco- in the shortening of APD or ERP and in the development
lemma, KATP channels are also present in mitochondria of atrial fibrillation. Interestingly, however, downregula-
(348, 349). Evidence suggests that both sarcolemmal tion of KCNJ3 gene coded mRNA, Kir3.1 channel protein,
and mitochondrial KATP play cardioprotective roles, albeit and IK,Ach and upregulation of microRNA (miR)-30d were
via different mechanisms (350). However, because of the also observed in atrial tissue obtained from patients with
lack of truly specific modulators for sarcolemmal and mi- atrial fibrillation (363). In this study, downregulation of
tochondrial KATP channels, this remains controversial and Kir3.4 protein was also reported (363), thus suggesting a
further research is needed. complex, microRNA-regulated ion channel expression in
atrial fibrillation that needs further investigation. In addi-
tion, after adenosine-induced atrial fibrillation, APD
3.8.2. Acetylcholine- and adenosine-activated shortened, more so in the right than in the left atrium,
potassium channels. and this effect was prevented by inhibition of IK,ACh with
the selective blocker tertiapin (364). Accordingly, the
It has been known for a long time that acetylcholine short- expressions of both adenosine receptor and Kir3.4
ens APD and has been implicated in atrial fibrillation (351). GIRK4 channel protein were different between the right
The ion channel that is directly affected by acetylcholine and the left atrium (365), thus suggesting a possible role
is called GIRK1/4 or Kir3.1/Kir3.4 channel (352, 353), of IK,ACh effect heterogeneity in evoking atrial fibrillation.
encoded by KCNJ3 and KCNJ5 genes, whose a-subunits Recently, it has been reported that an inherited gain-
are closely coupled to muscarinic M2- and adenosine A1- of-function mutation of KCNJ5 caused familial human
receptor proteins (354). These channels carry an inwardly sinus node disease. The enhanced activity of GIRK

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1101


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

channels was associated with maintained hyperpolariza- (376). It has been also demonstrated that TMEM16 and
tion of the pacemaker cells that resulted in reduced Bestrophin-3 are colocalized with Cav1.2 in canine and
heart rate (366). human left ventricular myocytes. In line with this, it was
found that activation of calcium-activated Cl current
3.9. Chloride Channels (ICl,Ca) requires Ca21 entry through sarcolemmal ICa,L,
and it is activated by Ca21 release from the SR.
Chloride or anion currents in cardiac muscle were Furthermore, ICl,Ca exerts an early repolarizing and a late
described long ago (367). However, despite intensive depolarizing component during the action potential,
and valuable research by some laboratories, relatively determined by 9-anthracene current in canine ventricu-
little attention has been paid to their possible role in car- lar myocytes (377).
diac electrophysiology and arrhythmogenesis (368). Despite the fact that ICl,Ca had been discovered a
One possible explanation is that the functions of chlo- long time ago in the heart, its genetic background was
ride channels are diverse (369, 370) and complex and not known for some time and was first attributed to the
their activation usually needs external triggers like bestrophin channel protein encoded by the VMD gene
enhanced intracellular cAMP, Ca21, and swelling or (378–380), even if later TMEM16 was reported as an al-
stretch (371). Also the research on Cl currents is ternative (381–383). The impact of this current on the
hindered by the lack of specific blockers, since the action potential is different from the other Cl currents
established inhibitors of these currents, e.g., DIDS, since it is strongly and dynamically dependent on the in-
9-anthracene (9-AC), tamoxifen, and Cd21, are not tracellular Ca21 concentration (375, 384, 385). It has an
specific. For these reasons, there are uncertainties important contribution to phase 1, early plateau repolari-
regarding the degree to which Cl channels affect dif- zation, and it also contributes to the repolarization
ferent types of cardiac action potentials. In the basal reserve. During Ca21 overload, when spontaneous
condition, they carry less current than other well- Ca21 release can happen during diastole, ICl,Ca can con-
established transmembrane ion channels. However, tribute to the development of DADs by carrying inward
in pathological conditions they may play a more im- current at voltages more negative than the Cl equilib-
portant role on arrhythmogenesis than previously rium potential (386). Recently, an anoctamin 1 (ANO1)-
thought. Therefore, Cl channels deserve more atten- encoded Ca21-activated Cl channel was identified in
tion, ideally in large-animal or human hearts. the ischemic heart, and its increased density was attrib-
So far, at least four Cl channels have been identified uted, at least in part, to the genesis of ischemia-induced
convincingly in the heart (369, 370). arrhythmias (387).
In contrast, in 5-day infarcted canine heart, the Ito2 cur-
3.9.1. Calcium-activated Cl channels. rent measured from myocytes of the epicardial border
zone exerted a significantly smaller peak amplitude,
It has been suggested that a current with properties sim- which may contribute to the development of an abnor-
ilar to Ito may be carried by Ca21-dependent chloride mal action potential (388).
and potassium currents (372, 373). In this context, it is
important to note that the first report of Ito in calf Purkinje 3.9.2. cAMP-dependent cystic fibrosis
fiber by Dudel et al. (372) attributed the current to a Cl transmembrane conductance regulator Cl
conductance. The kinetics of this Ca21-dependent Ito channels.
reflects the kinetics of the calcium transient or rather the
changes of intracellular Ca21 in the vicinity of the sarco- These channels are mostly closed under basal condi-
lemma. This current was also defined earlier as Ito2 or tion, and they carry outwardly rectifying Cl current
Ito,slow and could be abolished by depleting the intracel- only when intracellular PKA- and PKC-dependent
lular Ca21 store with caffeine (374) or directly inhibited phosphorylation is enhanced. Since the equilibrium
by DIDS or SITS (375) or by 9-anthracene (9-AC). To potential for Cl in normal conditions is between 65
avoid confusion, it must be emphasized that this current and 40 mV (389–391), they can carry both inward
should be distinguished from the Kv1.4 current, which and outward current during the action potential.
has also been referred to recently as slow Ito because of However, because of its outward rectification, the
its slow recovery from inactivation. The physiological main effect of the cystic fibrosis transmembrane con-
and pathophysiological roles of the Ca21-dependent Ito ductance regulator (CFTR) Cl current is to shorten
are not well defined but were suggested to play some APD and consequently ERP, which in turn may facili-
role in frequency-dependent APD regulation and to tate reentry arrhythmias. Interestingly, it was found
secure or shorten repolarization, possibly preventing or that the CFTR Cl current abolishes early and late
diminishing calcium overload in pathological settings preconditioning and also plays a role in

1102 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

postconditioning-related cardioprotection (392). It channels are constitutively active in cardiac hypertrophy


was also reported that intracellular Na1 modulated and heart failure (370, 371). This may limit APD prolonga-
the cAMP-dependent regulation of this channel (391). tion, but it may facilitate DADs at elevated diastolic
[Ca21]. In addition, since Ito and IK1 are downregulated in
the failing heart (7), this persistent ICl,swell may further
3.9.3. Swelling- and acidosis-activated CIC-2 Cl change the balance of inward and outward currents to-
channels. ward the inward direction, thus favoring membrane
depolarization (405). It was also reported that targeted
In cardiac myocytes, another Cl channel was also inactivation of the CIC-3 gene prevents the cardiopro-
described, encoded by the CIC-2 gene and carrying a tective effect of the late but not the early phase of pre-
current that is activated by hyperpolarization more nega- conditioning in mice (405).
tive than 40 mV, with a relatively slow biexponential
time course (393). The current is increased by swelling
3.10. Two-Pore Domain Channels
and acidosis, and it is blocked by 9-AC and Ca21 but not
by tamoxifen or DIDS (394). CIC-2 current, also called
Two-pore domain K1 channels (K2P) are widely exp-
ICl,ir, is strongly rectifying in the inward direction (370,
ressed in different organs, including the heart, but rela-
394, 395), and therefore it plays no or little role in car-
tively little is known regarding their function in the myo-
diac repolarization. However, it seems to be important in
cardium. Except for a few studies, the physiology of
the SA node (396), where it is abundantly expressed,
these channels has been studied only in mice or rats,
and may contribute to enhanced automaticity during is-
but they are abundantly expressed in the human heart
chemic cell swelling and acidosis.
as well (406, 407). In general, two-pore domain K1 chan-
There are some data regarding a strongly outwardly
nels represent a superfamily of large-conductance K1
rectifying acidosis-induced Cl current (ICl,acid), but its
channels, which lack voltage dependence or may ex-
molecular entity is unknown. It can be assumed that this
hibit outward rectification (64, 408). They are composed
current can contribute to the APD shortening in ischemia
of two a-subunits, each containing four transmembrane
(397).
segments that form two ion-conducting large-conduct-
Protein tyrosine kinase is one of the initial factors
ance pores (FIGURE 3), and may be associated with sev-
responding to cell swelling, providing the possibility that
eral types of accessory subunits (5, 64). These channels
tyrosine kinase is an important upstream regulator of the
are considered important determinants of background
swelling-activated Cl current (ICl,swell) (398, 399).
K1 conductance (408). They contribute to the resting or
Furthermore, angiotensin II is released during stretch,
maximal diastolic potential and to the repolarization
and it is implicated in cardiac remodeling and develop-
phase and are regulated by stretch, pH, temperature,
ment of HF, where ICl,swell is chronically activated (400,
and lipids or various signaling messengers (408). In gen-
401). Block of AT1 receptors prevented activation of
eral, the exact roles of the two-pore channels in cardiac
ICl,swell, thus suggesting a potential role of angiotensin II
electrophysiology and arrhythmogenesis are not well
in the activation of the current (402).
explored and offer challenging targets for further res-
earch. Therefore, future intensive research seems
3.9.4. Volume-regulated CIC-3 Cl channels.
worthwhile to clarify the function of these channels in
larger mammals and humans, including their potential
The channel encoded by the CIC-3 gene carries a time-
role in arrhythmogenesis.
independent and robust outwardly rectifying Cl current
(ICl,swell) that is small in basal isotonic conditions but is
increased by swelling or stretching (369, 370). During is- 3.10.1. TWIK-1 channels.
chemia and reperfusion, when swelling of myocytes can
occur, ICl,swell shortens APD and may enhance disper- The “two-pore” or “Tandem of pore domains in a
sion of repolarization between ischemic and nonische- weak inward-rectifying K1 channel 1” (TWIK-1) channel,
mic zones, thus facilitating both atrial and ventricular encoded by the KCNK1 gene, was first described in
fibrillation (371, 393). Since the CIC-3 channels are 1996 (407). They are expressed in the heart, are acti-
expressed in atria, ventricles, Purkinje fibers, and SA vated by mechanical stretch, intracellular acidification
node cells, they could also play a role in mechanotrans- (409), and polyunsaturated fatty acids, and facilitate
duction and in normal pacemaker function, and they repolarization and membrane hyperpolarization (5).
may contribute to the development of extrasystoles by Recently, it has been reported that TWIK-1 channels
promoting membrane depolarization in both the atria can change selectivity from K1 to Na1 under extrac-
and ventricles during stretch (403, 404). CIC-3 chloride ellular acidic conditions and low K1 concentration

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1103


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

(410), and this phenomenon is responsible for the 3.10.3. TREK-1 (K2P2.1) channels.
paradoxical depolarization contributing to enhanced
arrhythmic activity in hypokalemia (411). The “TWIK-related K1-channel” (TREK-1) represents
another two-pore K1 channel entity that is widely and
3.10.2. TASK-1 (K2P3.1) and TASK-3 (K2P9) abundantly expressed in various cardiac tissues (5, 419,
channels. 420). TREK-1 channels are particularly important since
they carry the outwardly rectifying K1 current activated
The “TWIK-related acid-sensitive K1” (TASK) channels, by stretch (420), the so-called stretch-activated cation
encoded by the KCNK3 and KCNK9 genes, respectively, current (SAC), temperature, or polyunsaturated fatty
are voltage-independent channels highly sensitive to acids, the latter also called arachidonic acid-sensitive
extracellular pH, and TASK-1 channels can be selectively current (IKAA). TREK-1-mediated SAC can shorten repola-
inhibited by A293 (5, 64, 412). TASK-1 channels can con- rization, by carrying outward current during the whole
tribute to APD shortening in ischemia because of their duration of the action potential, and hyperpolarize the
inhibition by extracellular acidosis (412). In addition, they membrane, thus in turn affecting spontaneous fre-
were shown to be upregulated in the atria of patients quency and impulse conduction. This current seems to
with chronic atrial fibrillation and preserved cardiac func- play a larger role in the atria and SA node than in the
tion (413), whereas a downregulation was recently ventricles. However, a pathophysiological effect known
observed in the atria of patients with atrial fibrillation and as “commotio cordis,” describing the accidental chest
reduced cardiac function (414, 415), associated with atrial hit that can cause sudden cardiac death, is presumably
APD shortening (412) and prolongation, respectively. In due to the function of TREK-1 channels in the ventricles
human atria, TASK-1 channels can form heterodimers (420). Interestingly, mutations in the selectivity filter can
with TASK-3 channels (KCNK9) that can be regulated by make the TREK-1 channels permeable to Na1, thus caus-
stress and thus contribute to the pathogenesis of ing ventricular tachycardia (421). TREK-1 channels are
chronic atrial fibrillation (416, 417). Interestingly, a gain- downregulated in atrial fibrillation and heart failure
of-function mutation in the TASK-4 (KCNK17) channel, (422–425). They also mediate cardiac fibrosis and dia-
which is sensitive to pH in the alkaline direction, was stolic dysfunction, via activation of c-Jun NH2-terminal ki-
reported recently (418), linked to a severe conduction nase (JNK) in myocytes and fibroblasts (426, 427), thus
disorder and suggesting the role of the TASK-4 channel enhancing the propensity of arrhythmias. Genes (Popdc
in both repolarization and membrane hyperpolarization- 1–3) were identified encoding the cAMP-binding Popeye
related conduction changes. protein, which associates with TREK-1 channels,

A AP prolongation
B C
Human &
Large mammal
Ventricle
ICa reactivation

FIGURE 10. Mechanisms of afterdepolari-


zation formation in cardiomyocytes. In ven-
0 mV
tricular myocytes from large mammals,
phase 2 early afterdepolarizations (EADs)
Phase-2 Phase-3 DAD
EAD EAD are associated with L-type Ca21 current (ICa,
) recovery from inactivation and reactivation
L
during prolonged action potentials (APs) (A).
Spontaneous sarcoplasmic reticulum Ca21
release, which increases Ca21 extrusion via
Na1/Ca21 exchange (inward current) (INCX),
can lead to phase 3 EADs (B) or delayed
INCX augmentation
afterdepolarizations (DADs) (C) when occur-
ring during or after the termination of repola-
rization, respectively. (Reproduced from Ref.
434 with permission.)

Spontaneous
Ca2+ release

1104 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

modifying their function. Accordingly, the absence of enters during the action potential (36). The pump
Popdc 1–2 genes should reduce TREK-1 current, increas- exchanges 3 Na1 for 2 K1 and thereby carries an out-
ing sinus node activity; however, the opposite has been ward current (INa/Kpump) (282). The size of the gener-
observed experimentally in mouse mutants (242, 428), ated current ranges between 0.34 and 0.68 pA/pF
suggesting the presence of yet unidentified interaction (37), which can result in RMP changes of 8–9 mV at
partners for Popeye proteins. Recently, it was proposed near-physiological conditions (35, 38), even though
that TREK-1 channels could also play a role in establish- its effect is often overlooked. It plays an important
ing the two levels of resting potential (429), a phenom- role in ionic homeostasis and also in cardiac repolari-
enon that had been described long ago in ventricular zation (435). The INa/Kpump shortens repolarization in
and Purkinje fibers (430, 431). simulations (436), contributes to the resting potential,
and, if its activity is enhanced by elevated extracellu-
3.10.4. Two levels of resting potential. lar [K1], intracellular Na1 concentration ([Na1]),
cAMP, or high firing rate (437), can even hyperpolar-
Because of the N-shaped steady-state current-voltage ize the cell (438). The Na1-K1 pump needs energy to
relationship of the Purkinje (431, 432) and ventricular work, since it pumps out Na1 against its electrochem-
(433) cells, two stable levels of resting membrane poten- ical gradient. The source of this energy comes from
tials can develop in these cells. The conductance of po- intracellular ATP, through the function of the Na1-K1-
tassium channels at 4 mM extracellular [K1] near the ATPase, which is part of the pump (438). So far, two
equilibrium potential for K1 is 100 times greater than a-subunits have been identified for NKA: a1 and a2
that of the Na1 and Ca21 background channels; there- (439, 440). The a1 is evenly distributed in the sarco-
fore it determines the resting potential to be around lemma (441), and it regulates intracellular Na1 in the
90 mV. When the conductance of potassium channels bulk of the cytoplasm. The a2 is mostly expressed in
decreases because of IK1 inward rectification (FIGURE 9) the dyadic cleft (440, 442, 443) and therefore may
and/or hypokalemia, the conductance ratio of K1 and have a special role in controlling, via interaction with
Na1/Ca21 channels can dramatically decrease, which the NCX, the Ca21 released from the sarcoplasmic
results in a second stable, albeit lower, potential in the reticulum (440). The fact that the a1-subunits of the
range of 50 to 25 mV depending on extracellular Na1-K1 pump are evenly distributed in the sarco-
[K1] or the actual strength of the background Na1 and lemma and carry >80% of the INa/Kpump (440, 444)
Ca21 currents (431–433). This second stable potential raises the possibility to develop drugs that selec-
has an important role in the genesis of the arrhythmo- tively modulate the Na1-K1 pump a1- and a2-subunits
genic triggered spontaneous automaticity like phase 2 (440). As well as being regulated by intracellular Na1
or 3 early afterdepolarizations (FIGURE 10). concentration (445), Na1-K1 pump activity is con-
trolled by the phosphorylation of FXYD protein phos-
pholemman (PLM) (446). Dephosphorylated PLM
3.11. Transmembrane Ion Transporters exerts a tonic inhibition of Na1-K1 pump, and its
phosphorylation would relieve this inhibition. Sti-
Transmembrane ion transporters, which exchange cati- mulation of the b-receptor increased the pump activ-
ons across the sarcolemma, are essential to maintain ity (447), and a-receptor activation enhanced the
the uneven ion distribution between the extra- and intra- Na1-K1 pump in canine Purkinje fibers (448). It was
cellular space. These ion pumps influence cardiac cellu- further reported that insulin flattened the pump cur-
lar electrophysiology indirectly, by changing various rent (Ip)-voltage (V) curve of the pump, depending on
intracellular concentrations (282), most importantly the patch pipette [Na1] and voltage (449).
Ca21, which activates and regulates several other trans- The b-subunit is also essential for pump function, provid-
membrane ionic currents. In addition, some transmem- ing a stabilization role for the a-subunit. The c-subunit
brane ion transporters are electrogenic, i.e., they seems to contribute in regulation of the sodium pump
exchange charges unevenly, thereby carrying inward or activity (450, 451).
outward transmembrane ion currents, which can influ- The cardiac glycosides (such as ouabain) are specific
ence the action potential repolarization and maximal dia- inhibitors of the Na1-K1 pump (452). Application of 1 μM
stolic or resting potential. ouabain increases the AP duration at 90% repolarization
(APD90) by 17% and 10 μM by 21%, also increasing the AP
3.11.1. The Na1-K1 ATPase/pump. plateau (453).
The Na1-K1 pump is a key player in the cellular mech-
The primary role of the Na1-K1 pump (NKA) is to anism of ischemia-reperfusion injury. In myocardial is-
remove from the intracellular space the Na1 that chemia, reduced ATP levels cause a decline in activity

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1105


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

for the Na1-K1 pump. The suppressed function of the augmented, 1 mM ORM-10962 moderately lengthened
pump leads to elevated intracellular Na1 level, which the action potential (469). It is generally considered that
activates reverse mode of the Na1/Ca21 exchanger NCX is the major, but not the only (472, 473), source of
(NCX), thus providing enhanced Ca21 entry and Ca21 Ca21 removal from the cell during the later phase of the
overload (4). action potential and diastole, carrying an inward current
that can contribute to both EADs and DADs (FIGURE
3.11.2. The Na1/Ca21 exchanger. 10). Indeed, it was demonstrated that inhibition of NCX
abolishes both EAD and DAD development (470, 474–
The NCX is a member of a large Ca21/cation antiporter 476) and also arrhythmias caused by enhanced disper-
superfamily (454). The mammalian tissues express three sion of repolarization (477) or by ouabain application
NCX isoforms, NCX1–NCX3, encoded by the genes (469).
SLC8A1 SLC8A3, respectively (455–457). Alternative Some studies suggest that NCX proteins are rela-
splicing of the primary nuclear SLC8A1 transcript gener- tively concentrated in the dyadic clefts, where the
ates at least 17 NCX1 proteins (454), and the NCX1.1 SR-ryanodine Ca21 release site faces the sarco-
splice variant represents the cardiac isoform of the NCX lemma (478–480), but another study (481) provided
(458). The NCX1.1 comprises 10 transmembrane seg- different results, and thus this issue is still unresolved
ments, where two groups of five transmembrane seg- (440, 463). Na1 entering the cell can increase the in-
ments are separated by an intracellular loop (459). This tracellular Na1 concentration in the small volume of
loop contains the exchanger inhibitory peptide (XIP) the dyadic clefts, thereby increasing Ca21 entry into
region that plays a key role in NCX inactivation by the cell on reverse-mode NCX (470, 482). This may
increased intracellular [Na1] (460, 461). The cytoplasmic increase Ca21-induced Ca21 release from the SR. It
loop also contains two calcium-binding domains that are is important to note that Ca21 concentration changes
responsible for Ca21 regulation of the NCX (462). The in the clefts and close to the sarcolemma can be
main function of the NCX is to control calcium flux orders of magnitude higher than in the bulk of the
through the plasma membrane, and it transports 3 Na1 cytosol (483), which is what is measured routinely
for 1 Ca21, using the driving force of the Na1 gradient with fluorescent Ca21 indicators. Therefore, experi-
provided by the NKA as discussed previously (458, ments can severely underestimate the electrophysio-
463–465). The stoichiometry makes NCX electrogenic logical role of NCX in cardiac myocytes. Further
(466), with one net positive charge moving either into methodological improvements are necessary to
the cell (“forward mode) or out of the cell (“reverse determine Ca21 concentrations most accurately in
mode”), eliciting inward or outward transmembrane cur- the dyads and clefts in order to better define and
rent (INCX), accordingly. The magnitude and direction of understand the electrophysiological role of NCX in
ion transport and generated transmembrane current arrhythmogenesis.
therefore depend on the membrane potential as well as Sorcin is a penta EF-hand protein that interacts with in-
the transmembrane concentration gradients of Na1 and tracellular target proteins after Ca21 binding. It has been
Ca21, thereby dynamically changing during the action reported that NCX1 could be an important target of sor-
potential (467). During the early phases of the action cin: downregulation of sorcin decreases NCX activity,
potential when transmembrane voltage is positive and but a higher level of sorcin increases it (484). It was also
intracellular Ca21 is low, Ca21 enters the cell by reverse described that insulin is able to increase the NCX activity
NCX, generating outward current. Later on, when [Ca21]i via interaction with the 562–670 f-loop domain (485).
is increased, Na1 enters and Ca21 leaves the cell, gen- Furthermore, the wild-type NCX1.1 associates with the F-
erating an inward current (468). This function results in actin cytoskeleton, probably through interactions involv-
complex changes on the cardiac action potential (467) ing the central hydrophilic domain of the NCX, and this
and arrhythmogenicity depending on heart rate, possi- association interferes with allosteric Ca21 activation
ble Ca21 overload, or NCX expression levels. It was (486). Phosphatidylinositol 4,5-bisphosphate (PIP2) plays
reported that a substantial degree of selective NCX inhi- an essential role in NCX regulation, since PIP2 increases
bition did not change the shape of the action potential reverse-mode NCX1 activity, causing a net increase in
waveform in normal physiological settings in dog ven- Ca21 influx (1133, 1134). Long-chain acyl-CoA esters,
tricular papillary muscle (469, 470) and in human right found to be elevated in cardiac ischemia (487), hypertro-
atrial preparations (471). However, when NCX forward phy (488), and failure (489), have been shown to be en-
mode was experimentally enhanced in dogs, selective dogenous activators of reverse-mode NCX activity by
NCX inhibition by 1 mM ORM-10962 shifted the plateau interacting directly with the XIP sequence, and thus link-
voltage toward negative values (469). On the contrary, ing altered fat metabolism to NCX function and NCX-
when reverse NCX mode was experimentally mediated calcium overload in the myocardium, in

1106 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

pathological conditions (490). Importantly, protons are (507) and cell volume (508). The function of NHE is
powerful inhibitors of NCX1.1 forward-mode activity, fur- electrically neutral; therefore, it does not directly
ther emphasizing the role of NCX1.1 as a key contributor affect the AP and arrhythmogenesis in normal condi-
to pathologically altered Ca21 homeostasis and arrhyth- tions. However, it has been shown to play important
mia generation during cardiac intracellular acidosis, pathophysiological roles by indirectly changing in-
including myocardial ischemia (491, 492). tracellular Na1 concentration and pH (509, 510),
The effects of genetic mutations influencing NCX both affecting several ion channels and transport-
function are poorly understood. However, a genetic ers. The role of NHE is considered significant in
mutation in NCX1 was demonstrated to cause cardiac fi- arrhythmogenesis in pathological settings, including
brillation in a zebrafish model (493). myocardial ischemia-reperfusion, heart failure, or di-
abetes, when intracellular pH or Na1 deviates from
3.11.3. The Na1/H1 exchanger. physiological levels (496, 511–514).

The Na 1/H1 exchanger (NHE) was first described in


rat intestinal and kidney tissue (494). Since then, 4. TISSUE-SPECIFIC ACTION POTENTIALS
nine NHE isoforms have been identified (NHE1 to
NHE9, encoded by the genes SLC9A1 to SLC9A9, 4.1. Sinoatrial Node and Pacemaker Function
respectively) (495, 496), and in the plasma mem-
brane of mammalian myocardial cells NHE1 is the The sinoatrial node (SAN) is located in the upper part
primary isoform (497). The NHE1 consists of 12 trans- of the right atrium, and it has a special role in the
membrane segments, with the NH2 terminal (500 heart, serving as the natural pacemaker. SAN cells
residues) catalyzing ion transport and interacting have a relatively low maximal diastolic potential (less
with pharmacological NHE inhibitors and the COOH than 60 mV), that, after the termination of repolari-
terminal (300 residues) responsible for regulation zation, gradually becomes less negative during the
of the exchanger by calmodulin, PIP 2, and calci- so-called spontaneous diastolic depolarization—until
neurin B homologous proteins (498–504). The NHE1 it reaches the potential range of L-type Ca21 channel
can be found as a homodimer in the plasma mem- activation, thereby generating a new action potential.
brane (505). The NHE1 dimer exchanges 2 extracel- The exact nature of the pacemaker function in the
lular Na1 for 2 intracellular H1 in one cycle (506), SAN is still under debate (515–519). Originally, it was
and it is an important regulator of intracellular pH thought that the slow diastolic depolarization was

0
3
20 mV MDP
4
DD
200 ms

ICaT ICaL ICI


IKr IK(ACh) If INCX Membrane-clock

Ca2+ Cl– K+ K+ Na+ 3 Na+ Ca2+ 2 K+ 3 Na+

Spontaneous,
rhythmic Ca2+ Ca2+ -clock
releases

FIGURE 11. Hypothetical mechanism for sinus node pacemaking. The figure illustrates a typical sinus node action potential (red trace) and the timing
of the membrane and Ca21 clock components. Upon phase 0, the T (ICa,T)- and L(ICa,L)-type Ca21 channels (and presumably the Cl channels, ICl) open,
providing inward current (downward arrows), and depolarize the membrane. During repolarization (phase 3) the opening of the delayed rectifier (I )
Kr
and the acetylcholine-dependent (IK,ACh) K1 currents (upward arrows) repolarizes the membrane to reach the maximal diastolic potential (MDP). Upon
diastolic depolarization (DD), the cAMP-dependent “funny current” (I ) slowly depolarizes the membrane in close cooperation with the sodium-calcium
f
exchanger (I ) that is governed by spontaneous, rhythmic Ca21 oscillations from the sarcoplasmic reticulum.
NCX

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1107


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

due to a hyperpolarization-activated inward current. studies suggested that mitochondrial Ca21 transient decay
This current was first described in cardiac Purkinje is slow, and that their Ca21 efflux is relatively small during
fibers and was thought to be carried by Na1, based one cardiac cycle (463), thus raising questions about a sig-
on its dependence on extracellular [Na1] (520). Later nificant role of mitochondrial Ca21 release on SAN fre-
on, other studies suggested that the slow diastolic quency. Further studies in adult SAN cells are necessary to
depolarization was the result of a decaying K1 cur- confirm the possible role of mitochondria in pacemaking.
rent called IK2 (521, 522). These data were subse- In general, SAN frequency can be slowed down by in-
quently reinterpreted in terms of ionic currents hibiting If, NCX, ICa,L, or IKr in feline, rabbit, and porcine
resulting from extracellular accumulation of K1, and a SAN myocytes (537–540). Inhibition of If and NCX
hyperpolarization-activated inward current, called decreases the slope of diastolic depolarization, while in-
“funny current” (If), carried by both Na1 and K1, was hibition of ICa,L would shift the threshold potential toward
described by Di Francesco (524) and others (523, more positive values, and thereby the cycle length is
525). Despite the fact that other currents, such as IKr, increased in all cases. Inhibiting IKr prolongs repolariza-
T-type ICa and NCX, were also suggested to play a tion of SAN cells and lengthens their spontaneous cycle
role in the SAN pacemaker activity (526–529), for lengths. It has to be considered, however, that loss-of-
more than two decades since its discovery the major function mutation of If results in sinus bradycardia, argu-
mechanism underlying the pacemaker function was ing for some role of If in SAN pacemaking (541).
generally considered to be If. However, in an early SAN frequency can also be affected by intracellular
study of Noma, Morad, and Irisawa (530), the signifi- cAMP levels. Elevation of intracellular cAMP increases If
cance of If in the SAN pacemaker function was ques- and shifts its activation to more negative potentials
tioned. These authors showed that cesium, an (542), which in turn enhances ICa and intracellular Ca21,
inhibitor of If, did not influence the SAN spontaneous further favoring the increase of SAN frequency. Vagal
frequency and did not eliminate the epinephrine- stimulation has an opposite effect on intracellular cAMP
induced increase in SAN frequency. The dominant and SAN frequency. Data from SAN cells isolated from
role of If in SAN pacemaker function was later also Girk4 (Kir3.4)-knockout mice lacking IK,ACh suggest that it
challenged by studies showing that a cycling sponta- may also activate IK,ACh, which can cause hyperpolariza-
neous release of Ca21 happens during diastole, and tion and may also contribute to SAN bradycardia (543).
this can cause spontaneous depolarization by acti- SAN cells lack cardiac type Nav1.5 fast Na1 channels,
vating forward NCX as an alternative mechanism for and therefore their depolarization is caused by ICa,L
the pacemaker function (“calcium clock hypothesis”) (544). Recently, neuronal Nav1.6 Na1 channels were
(203, 531–534). This controversy seems to be settled reported in SAN cells (90, 545), but their role in SAN
by the “coupled clock hypothesis” (FIGURE 11), sug- function is not well understood (529). The SAN action
gesting an important role for both the “calcium clock” potential does not show a distinct plateau phase with
and the “membrane clock” (If) (528, 535). Also, in a relatively weak IK1 current.
recent study, Morad and Zhang (517) demonstrated Experimental evidence obtained in spontaneously
and pointed out that If was very small and very slowly beating guinea pig sinoatrial cells suggests that If function
activating at the range of maximal diastolic potential decreases SAN frequency variability, which is the intrinsic
of the SAN cells ( 60 mV), therefore not enough to behavior of the calcium clock function (546).
generate a significant amount of pacemaker current. Elucidating the exact mechanisms underlying pace-
These authors suggested that expression and func- maker function and its regulation still remains an impor-
tion of inward If in SAN cells can counterbalance the tant task for the future, since it provides the regular
electrotonic interaction of the more negative resting heartbeat but it can also act as a possible trigger for seri-
potential of the surrounding atrial cells. According to ous atrial and ventricular arrhythmias (547, 548).
this suggested function, If is important since it insu-
lates SAN cells from the hyperpolarizing influence of
the atria, thus allowing a proper SAN function. 4.2. Atrial Action Potential
Consistent with this speculation, Boyett et al. (536)
found higher HCN (If) expression in peripheral rabbit Large species-dependent variations make it difficult to
SAN tissue than in the central core. describe the general shape of cardiac action potentials,
A recent study in human induced pluripotent stem cell including atrial action potentials (FIGURE 12 and
(HiPSC)-derived myocytes suggested that mitochondria FIGURE 13). There is also significant diversity within the
could also play a role in the spontaneous activity, setting same heart (549), at least in humans (3, 550), and this
the rhythm of the “calcium clock” by taking up and releas- results in a relatively large dispersion of repolarization,
ing Ca21 from and to the cytosol (517). However, other which favors the development of atrial fibrillation.

1108 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

FIGURE 12. Species-dependent differences in atrial (A) and ventricular (B) action potential configuration. Original action potential recordings at 1 Hz
stimulating frequency from human, dog, rabbit, guinea pig, and rat ventricular muscle preparations recorded by the conventional microelectrode tech-
nique. Unpublished data from our laboratory at the Department of Pharmacology and Pharmacotherapy, University of Szeged.

In most species, the atrial action potential lacks a long compared with normal conditions. It is interesting that, in
and stable plateau phase. In human atria, most of the rabbit (unlike in human) atrial muscle, sustained Cl cur-
atrial action potential recordings show a plateau phase, rent was reported after Ito inactivation (324, 553). This
but at a more negative voltage range ( 10 to 30 mV) should be taken into consideration when drugs are stud-
than observed in the ventricle. This is due to the function ied in rabbit atrial preparations, and it highlights the im-
of the abundantly expressed Ito and IKur potassium cur- portance of species-dependent electrophysiological
rents (FIGURE 6). Ito in the atria has slower inactivation differences. A distinct, cellular swelling-induced Cl cur-
kinetics than in the ventricles, having a robust slowly rent has also been described in human atrial myocytes
deactivating component (s = 91 ms at 20 mV), with (554), which is also modulated by PKA-independent,
slower recovery (551) from inactivation (s = 125 ms) than cAMP-mediated b-adrenoceptor signaling (555). The ex-
the ventricular one. Therefore, frequency-dependent istence and potential role of Nav1.8 channels in INaLate in
changes of APD and restitution are different in atrium the atria represents an interesting issue; however, it is
and ventricle. In the atria IKur is large (85) and contributes still the subject of debate (556) and requires further
to repolarization, in contrast with the ventricle, where it studies. L-type and T-type ICa have similar properties in
is absent or very weakly expressed (85). Therefore, IKur atria and ventricle (557). IK1 current density is relatively
inhibition would be expected to prolong the AP. small in the atria, especially in the voltage range
However, inhibition of IKur shortens repolarization by between 80 and 0 mV. This is consistent with the find-
shifting the plateau voltage to the positive voltage range ing of lower Kir2.1 mRNA expression in right atrial com-
(309), thus changing the activation and deactivation/ pared with right ventricular human tissues (85). Recently,
inactivation of other plateau currents, such as IKr, IKs, ICa, it has also been reported that neurokinin-3 receptor acti-
and INaLate. IKur inhibition has also been shown to slightly vation induced prolongation of atrial refractoriness,
prolong human atrial APD in tissue obtained from which was attributed to the inhibition of a nonspecific K1
chronic AF patients (309, 552). However, in this case, background current (297). It is difficult to measure IKr
phase 1 and 2 repolarization were delayed and shifted and IKs in atrial myocytes, most likely because of the cell
to positive potentials, because of electrophysiological isolation techniques (252). Regardless of that, IKr block
remodeling, thus allowing for a larger IKur contribution significantly lengthens atrial repolarization, in both

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1109


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

FIGURE 13. Distinctly different effects of slow component of delayed rectifier potassium current (I ), transient outward current (I ), and ultrarapid
Ks to
component of delayed rectifier potassium current (IKur) inhibition on ventricular repolarization in human (top) and rat (bottom). In these experiments,
action potentials were recorded in ventricular papillary muscle with the conventional microelectrode technique at 1 Hz stimulation frequency. Note the
extended timescale of the recordings in rat papillary muscle. A: rapid component of delayed rectifier potassium current (IKr) inhibition by 50 nM dofeti-
lide markedly prolonged action potential duration (APD) in human but not rat papillary muscle. B: I block by 100 mM chromanol 293B in human elimi-
to
nated the notch and elevated the plateau potential to positive direction without changing the APD; however, the APD was significantly prolonged in
rat. Since chromanol 293B fully blocks IKs in addition to inhibition of Ito at 100 mM, prior IKs block was elicited by 500 nM HMR-1556 to dissect these
effects (140). C: I inhibition by 3 mM XEN-D0101 (310) does not affect APD in human but markedly prolongs APD in rat ventricular papillary muscle
Kur
(unpublished experiments from the Department of Pharmacology and Pharmacotherapy, University of Szeged).

animal and human studies (558, 559). On the other 562). The increased apamin-sensitive SK current was
hand, the role of IKs in atria is not well explored. found along with decreased mRNA and protein levels of
Experiments with chromanol 293B on atrial action SK1, SK2, and SK3 channels in human atrial cardiomyo-
potential are not conclusive, since this drug inhibits both cytes isolated from patients with AF (319). However, in the
IKs and Ito (560). It must be emphasized that, in physio- same experiments CaMKII was increased and its inhibi-
logical conditions, the atrial plateau voltage is more neg- tion by KN-93 reduced the apamin-sensitive SK currents
ative than the activation threshold of IKs. Therefore, IKs is to a higher degree in myocytes isolated from patients
not expected to contribute to atrial repolarization. with AF compared with those in sinus rhythm (319), sug-
However, it has been reported that in atrial tissue LQT1, gesting that SK channels are more sensitive to Ca21 in
MinK, and MiRP levels are similar to those in ventricular AF patients and CaMKII modulation may represent a
tissues (85). On the basis of these results, it can be pharmacological target in the management of AF.
speculated that, at fast heart rate (enhanced sympa- Neurohor-monal modulation of the atria is particularly im-
thetic tone) and in situations where the atrial plateau is portant. Acetylcholine (358, 563), catecholamines (555,
shifted to positive voltage, IKs may have a role in atrial 563), substance P (297), adenosine (564, 565), and sero-
repolarization. Therefore, its modulation could influence tonin (566) have been reported to influence atrial action
arrhythmogenesis. Indeed, it has been shown that two potential and its underlying currents. In the atria, IK,ACh is
gain-of-function mutations in KCNQ1 (S140G and V141M), robust and even has a small but persistent constitutively
detected in patients with AF (275, 277), markedly slowed active component, which operates without parasympa-
deactivation of IKs and contributed to the development thetic stimulation and seems greatly augmented during
of AF (561). Unlike in the ventricles, most studies showed chronic AF (358). Accordingly, atrial tissue expresses
the existence and function of small-conductance, apa- mRNAs for Kir3.1 and GIRK channels abundantly. Of note,
min-sensitive and calcium-dependent potassium current TWIK TASK channels are also relatively abundantly
(SK2) in normal atria, but its significance seems to be far expressed in the atria (85), but their roles in atrial electro-
more pronounced in diseased tissue (5, 314, 316, 317, physiology are not well explored yet.

1110 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

A left atrium pulmonary vein smooth muscle

Membrane Potential

Membrane Potential

Membrane Potential
50 50 50
25 25 25
0 0 0

(mV)

(mV)

(mV)
-25 -25 -25
-50 -50 -50
-75 -75 -75
-100 -100 -100

0 1000 2000 3000 4000 0 1000 2000 3000 4000 0 1000 2000 3000 4000

Time (ms) Time (ms) Time (ms)

B control
before stretch 30s after stretch

-50

1s 1s

gadolinium
before stretch 30s after stretch

-50

1s 1s

C
0.25 s

mV

-50

FIGURE 14. Properties of action potentials from left atrial and pulmonary vein myocytes. A: recordings of action potentials from the left atrium and pulmonary
vein and a transmembrane potential in the smooth muscle cell layer. (Reproduced from Ref. 576 with permission.) B: effects of mechanical stretch on the sponta-
neous electrical activity of the pulmonary vein myocardium. Expanded traces obtained before (left) and 30 s after (right) the application of mechanical stretch
(100 mg) in the absence (top) and presence (bottom) of gadolinium. (Reproduced from Ref. 577 with permission.) C: spontaneous action potential with early after-
depolarizations in canine pulmonary veins from dogs subjected to chronic atrial tachypacing. (Reproduced from Ref. 578 with permission.)

4.3. Pulmonary Veins (571, 572), to understand their role (573, 574), the mech-
anisms, and possible treatment of arrhythmias initiated
The spontaneous activity of the pulmonary vein (PV) was and maintained by the PVs. It was demonstrated that tis-
described long ago by several investigators (567, 568). sue with myocardial origin is present in the PVs of dogs
Later, Haïssaguerre and colleagues (569, 570) demon- and guinea pigs, with electrophysiological properties
strated that electrical activity in the pulmonary vein that are similar to but also distinct from the neighboring
sleeves (PVs) in patients has an important role in atrial fi- atrial tissue (568, 575) (FIGURE 14). PVs cells have less
brillation. In the following years, intensive research was negative resting potential and shorter APD than atrial
carried out both in the clinical and experimental fields myocytes and also lack a prominent plateau phase

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1111


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

(FIGURE 14). Work on canine PVs found that these cells frequency and a more negative maximal diastolic poten-
either do not show spontaneous diastolic depolarization tial (596, 597). The structure of the AV node, like the
or, if they do, their rate is normally lower than that of SAN, includes a large variety of different cell types (598,
SAN cells (576, 579). Stretch can also increase automa- 599), with different channel expression (600–603). In
ticity and induce high-frequency firing in guinea pig PV the rat AV node, high expression levels of HCN4, Cav3.1,
cells (577) (FIGURE 14). Sympathetic stimulation or dis- Cav3.2, Kv1.5, Kir3.1, and Kir3.4 and low expression of
eases like hyperthyroidism (580) increase intracellular Nav1.5 and Kir2.1 mRNA were measured compared with
[Ca21], causing PVs cells to develop triggered activity those observed in the ventricle (600). In rabbit AV node,
(FIGURE 14), and both early (EAD) and delayed (DAD) no or low expression of Nav1.5, Cav1.2, Kv1.4, KChIP2,
afterdepolarizations can occur (578) (FIGURE 14). and RYR3 and high expression of Cav1.3 and HCN4
Compared with atrial myocytes, PVs cells have similar mRNA were reported (601). Like SAN cells, AV nodal
ICaL, ICaT, Ito, and NCX but lower current density of IK1, IKr, cells do not have functioning fast Nav1.5 INa channels,
and IKs, with inconsistent results available regarding INa and therefore their depolarization and impulse conduc-
(576, 581). Interestingly, a hyperpolarization-activated tion depend on the function of ICa,L (565, 603). In AV
inward K1 current was reported in canine PVs cells, which nodal cells, the expression and function of IK,ACh is par-
was highly sensitive to low submicromolar concentration ticularly important (604, 605), since their activation via
of Ba21 (579). Based on this finding, Kir3 subunits were the adenosine 1 or muscarinic receptors (606) hyperpo-
speculated to be involved as the channel background of larizes the AV nodal cells, which slows or blocks impulse
this current, and it was suggested that it may play a signifi- conduction in the AV node (607, 608). In addition, aden-
cant role in atrial pathophysiology (579). This current, IK, osine and acetylcholine decrease ICa,L via Gi protein sig-
ACh, was enhanced in a conscious tachypaced dog AF naling pathway, further decreasing the safety of impulse
model, and its inhibition by tertiapin-Q resulted in a propagation through the AV node. These are the princi-
marked reduction in the incidence of AF episodes (558). pal cellular mechanisms that make intravenous adeno-
In rat PV, a voltage-dependent Cl current sine so useful in stopping AV nodal reentry tachycardia
was also reported and suggested to contribute to (604). The T-type Ca21 channel is expressed and is
norepinephrine-induced automaticity in the PVs. (582). functional in the AV node as in the SAN (607). Indeed,
PVs electrophysiology is also modulated by a wide variety mibefradil, a potent ICa,T inhibitor, increased AV nodal
of drugs that are used in clinical practice including cele- conduction time and even elicited second- or third-
coxib, amiodarone, ranolazine, losartan, and enalapril degree AV block in isolated, blood-perfused dog hearts
(583–587). Several factors and diseases, e.g., stretch (607). The slow conduction through the AV node is
(585), electrolyte disturbances, sex differences, air pollu- physiological and provides a time lag for contraction
tants (H2S) (588), thyroid hormone (580), ischemia-reperfu- between the atria and the ventricles. AV nodal tissue
sion (589), uremic substances, renal and heart failure, and has diverse and different connexin 40, 43, and 45 distri-
aging, have been reported to influence the electrophysiol- bution compared with other parts of the heart (8), with a
ogy of PVs. complex and diverse structure, containing a dual faster
Recently, it was shown that factor Xa inhibitor anticoa- and slower impulse conduction pathway (594, 603,
gulants, which reduce the incidence of AF-related 609). This latter can provide the basis of fixed-rate
stroke, also decrease the activity of rabbit PV cells, by in- supraventricular reentry tachycardia as was elegantly
hibiting PAR1 and also diminish INaLate. Based on this, it demonstrated in an early study in rabbits by Janse et al.
was speculated that they can modulate the occurrence (610), and this tachycardia can be terminated by block-
of atrial fibrillation (590). ing ICa,L and IK,ACh and by adenosine (604, 611, 612).
Although it is generally accepted that PVs represents AVN cells lack Ito and have background sodium inward
an important trigger for arrhythmias, further research in current flowing through a nonselective cation channel
this field is still necessary to fully elucidate the role of PVs (605, 613). These cells express functioning IKr, IKs, and If,
in arrhythmogenesis and to develop new effective thera- but If is not required for their pacemaking (614).
pies involving PVs in general (586, 587). Neverthe-less,
PV isolation by catheter ablation has become an estab- 4.5. Purkinje Fiber Action Potential
lished therapeutic intervention in the management of par-
oxysmal and persistent AF (591, 592). Purkinje fibers play a pivotal role in impulse conduction
and propagation in the ventricles (615, 616). Purkinje
4.4. AV Nodal Action Potential and Conduction cells can also act as subsidiary pacemakers, and they
display a spontaneous diastolic depolarization, although
Although they have similarities, AV node cells differ from their frequency is normally inhibited by the higher-fre-
the SAN cells (593–595), with a lower spontaneous quency discharges of the SAN, causing overdrive

1112 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

suppression. Purkinje fibers have a longer APD than do whereas in human Purkinje fibers abundant Kv3.4 and
ventricular cells, and this may have a protective function Kv4.3 expressions were reported, with marked differ-
against retrogradely propagating stimuli (615) but it can ences also in b-subunit (KChIP2, KChaP, KCNE2)
also represent a source of arrhythmogenic repolariza- expression patterns (158). The slow Ca21-dependent
tion inhomogeneity, even in the normal heart, which can Ito2 Cl current was also described in rabbit Purkinje
increase in diseased conditions or after drug exposure. fibers (631) and implicated in phase 1 repolarization
In certain pathophysiological situations, Purkinje fibers and DAD formation in sheep Purkinje cells (632), since
can show triggered activities (617), such as EADs, DADs, at potentials more negative than the chloride equilib-
and cellwide ectopic Ca21 waves, in surviving tissue in rium potential chloride channels conduct depolarizing
the border zone of an infarct (618). It was recognized current. The L-type ICa is well expressed in Purkinje
long ago that Purkinje strand fibers, which run close to fibers and, unlike in ventricular myocytes, it is carried
or on the surface of the endocardium, are less affected not only by Cav1.2 but also by Cav1.3 channels (633),
by ischemia-induced tissue damage compared with ven- and this may result in some differences in the proper-
tricular cells (118, 619). Purkinje fibers have been exten- ties of ICa,L between ventricular and Purkinje myocytes.
sively studied with the conventional microelectrode In the canine Purkinje fiber, there is larger T-type ICa,
(620) and the two-electrode voltage-clamp (621) techni- with a higher Cav3.2 expression than in ventricular and
ques. However, since cell isolation from cardiac Purkinje atrial myocytes (633). Based on this, it was speculated
tissue is particularly difficult, research in Purkinje fibers that T-type ICa has an important role in Purkinje fibers,
has benefited less from the introduction of the patch- by contributing to both depolarization and pacemaker
clamp technique, despite its importance (622). function (634). IKr, IKs, calcium-activated K1 currents, as
Purkinje fibers have a special role in impulse con- well as IK1 have been described in Purkinje fiber (323,
duction, with their depolarization being about two to 635, 636). Accordingly, inhibition of IKr and IK1 signifi-
three times faster (300–750 V/s) than in atrial or ven- cantly lengthens repolarization, even more so than in
tricular (100–250 V/s) muscle preparations (623). The the ventricular muscle (615). However, the inhibition of
fast depolarization is thought to be due to the abun- IKs does not change repolarization of Purkinje fibers in
dant expression of the TTX-sensitive Nav1.5 channel the normal situation (256). This is not surprising, since
isoform (85), but low density of TTX-sensitive neuronal Purkinje fibers have a plateau voltage less positive
Nav1.1 and 1.2 Na1 channel isoforms may also signifi- then 0 mV, which is below the activation threshold of
cantly contribute to INa in Purkinje fibers (624, 625). IKs. However, at high frequencies and a high level of
Importantly, the relation between impulse conduction, sympathetic activation, the plateau level is shifted to-
upstroke velocity (Vmax), and ionic currents is not linear ward more positive values, increasing both IKs ampli-
(626–628), and Vmax is not a direct measure of ionic tude and speed to such a level to influence both
currents. INa in Purkinje fiber also seems to have a par- repolarization and pacemaker function (256).
ticular impact on repolarization, due to its relatively Unlike ventricular but similarly to atrial muscles,
large slowly inactivating component (12, 13, 82). Some Purkinje fibers express IK,Ach and Kir3.1 GIRK channel
authors suggested that the skeletal muscle Na1 chan- subunits, thus responding with an APD shortening
nel isoform Nav1.4 (78) while others suggested that upon acetylcholine administration (637). The pace-
Nav1.7 subunits (85) were responsible for this slowly maker If current is robust in Purkinje fibers (638–640),
inactivating component in Purkinje fibers; however, where it was first discovered (524), and it plays an im-
this issue is not clarified and needs further investiga- portant role in the pacemaker function. Later, a K1 cur-
tions. In canine Purkinje fibers, connexin 40 is more rent, IKdd, was also described in Purkinje fibers (640),
abundantly whereas connexin 43 is similarly deactivating at more positive potentials than If and
expressed compared with ventricular myocardium thus having an additional role in the pacemaker func-
(629): this can play a role in differences between their tion in Purkinje fibers. In addition, spontaneous Ca21
conduction properties. The large phase 1 repolariza- release-induced intracellular Ca21 waves can also
tion in Purkinje fibers (FIGURE 6) is caused by the fast modulate normal Purkinje fiber pacemaker activity
inactivation of INa and activation of Ito. Not only is Ito (641). This may have particular importance in ectopic
larger than in ventricular muscle, but it inactivates automaticity of Purkinje fibers surviving myocardial in-
somewhat slower and recovers from inactivation >10 farction (618). Recently, significant SK2 current and
times slower than that in the ventricular muscle cells channel expression were described in rabbit Purkinje
(630). This behavior of Ito is attributed to the different fibers, and an important role for SK2 current in Purkinje
expression of Ito a- and b-subunits in dog ventricular fiber repolarization was suggested (323).
muscle and Purkinje fibers (141, 157). In human ventricu- Free-running Purkinje strands emerging distally into
lar myocytes Kv4.3 channel subunits dominate, ventricular muscle constitute a relatively large-

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1113


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

FIGURE 15. Quinidine-induced early afterdepolari-


zations (EADs) in canine Purkinje fibers propagate to
the ventricular myocardium. Top: multiple EADs origi-
nating from Purkinje fiber (ME1) and propagating to
the ventricular myocardium, recorded at position
ME2. Bottom: experimental setup and the positions
of the 2 microelectrodes (ME). (Modified from Ref.
643 with permission.)

resistance connection (642), and a high degree of sink 4.6. Ventricular Action Potentials: Transmural and
for current flow, and a more favorable site of conduction Regional Differences
block than other parts of the healthy myocardium. Also,
because of the weaker electrotonic coupling, the disper- Ventricular action potentials (FIGURE 6) in humans and
sion of repolarization here can be far greater than in in most mammals have a positive (120 to 130 mV) and
other places (201). Indeed, it was experimentally proven relatively long plateau phase, with a small or pro-
that EADs in free-running Purkinje strands could elicit nounced phase 1 repolarization and notch afterwards,
extrasystoles in ventricular muscle (643) by electrotonic depending on their transmural site of origin (subendo-
interaction (FIGURE 15), which is unlikely to happen in cardial, midmyocardial, or subepicardial) (143, 645, 646).
healthy and well-coupled regions within the ventricular Regardless of their origin, they express large INa and
wall. ICa,L and relatively weak but persistent INaLate (80). These
Since Purkinje fibers are considered particularly im- currents provide robust depolarization and thereby
portant in arrhythmogenesis (644), further research secure impulse conduction. In addition, by counterbal-
studying Purkinje fiber ventricular muscle junctions or ancing outward potassium currents, e.g., Ito, IKr, IKs, and
subendocardial layer containing a mixture of Purkinje IK1, they participate in the maintenance of the plateau
fibers and ventricular muscle would offer promising phase (FIGURE 6). As mentioned above, IKur and IK,Ach
results. are expressed weakly or not at all in the ventricles.

1114 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

There are several reports indicating electrophysiolog- considered a contributor to ventricular tachycardia and
ical differences between the right and left ventricle. In fibrillation development in patients with Brugada syn-
dog hearts, it was found that left ventricular myocytes drome, acquired and congenital LQT syndromes, short
had longer APD than those in the right ventricle (647, QT syndrome, and catecholaminergic polymorphic ven-
648). Also, right ventricular myocytes exhibited more tricular tachycardia (CPVT) (656). However, these differ-
pronounced phase 1 repolarization and larger Ito (648, ences in transmural repolarization are relatively small in
649) and increased IKs (648). It was also reported that the intact, undiseased ventricles (642, 661), since the
canine subepicardial and subendocardial myocytes in neighboring myocytes are well coupled. Accordingly,
the left ventricle possessed larger INa with higher Vmax experimental evidence suggests that in the intact heart
compared with those in the right ventricle (650). The the transmural dispersion of repolarization is consider-
authors suggested that these differences provided a ably less (642, 661, 662) than that reported in tissue sli-
mechanism for the right ventricular manifestation of ces and perfused wedge preparations (663, 664).
Brugada syndrome (650). Ther-efore, it was argued that it is unlikely that a large,
Almokalant, a drug with APD-prolonging effects, significant repolarization gradient between the sube-
increased interventricular dispersion of repolarization ndocardium, M cells, and subepicardium existed, con-
that was associated with the occurrence of EADs and tributing to EADs or arrhythmias (665). However, in
torsade de pointes arrhythmia in dogs with chronic AV pathological settings and/or drug treatment and at
block (647). In these dogs, the APD prolongation was Purkinje fiber ventricular junctions (FIGURE 15), APDs
larger in the left ventricle than in the right ventricle (647). can lengthen in a nonuniform manner and cellular
The opposite was observed in guinea pigs, i.e., the APD- coupling can deteriorate, thus resulting in a substrate
prolonging drugs dofetilide and quinidine lengthened for serious ventricular arrhythmias.
APD more in the right ventricle (651). These findings Apico-basal electrophysiological differences have
highlight important differences among species in their also been described: the APD was shorter and phase 1
responses to drugs with repolarization-prolonging effects. repolarization was markedly larger in canine cardiomyo-
In humans, similarly to dogs, the APD is longer in the left cytes isolated from the apical region than in those from
ventricle with slower adaptation to increased heart rate the basal region (655). In the same study, larger Ito and
than in the right ventricle (652). These interventricular dif- IKs were observed in apical than in basal cardiomyocytes
ferences in repolarization are not sufficient to cause (655). In this context, the apico-basal and the antero-
arrhythmias in the normal heart; however, in the presence posterior, and not the transmural, repolarization gra-
of an ischemic region at the left ventricle (LV)-right ventri- dients have been considered to contribute to the gener-
cle (RV) junction, the interventricular APD heterogeneity ation of the T wave (665, 666).
and different AP rate adaptation promote reentry arrhyth- Since idiopathic ventricular arrhythmias often origi-
mias (652). nate from the right and left ventricular outflow tracts
Important regional electrophysiological differences (RVOT and LVOT), the electrophysiological properties of
were described in the dog ventricle long ago (653), these regions have also been investigated (667–670). It
transmurally (654), regionally (655), and in the basoapi- was found that rabbit right ventricular myocytes from the
cal direction (143). These differences are due to substan- apex had longer APD, larger Ca21 transients, higher
tial differences in the density of expression of various Ca21 stores, increased INaLate and Ito, but smaller IKr, L-
transmembrane ion channels, shaping the action poten- type Ca21 current, and NCX than RVOT myocytes (671).
tials accordingly in the different regions of the ventricles These differences were associated with increased inci-
(645). Transmural differences in repolarization have dence of DADs induced by pacing (671). Myocytes from
been extensively studied and are well explored (656). It the LVOT exhibited longer APD, larger INaLate and NCX,
has been found that subepicardial cardiomyocytes ex- and smaller Ito and IKr than those from the RVOT (669).
hibit a large phase 1 repolarization and Ito compared
with those in the subepicardium (657). Myocytes iso- 4.7. Species-Dependent Differences in Action
lated from the midmyocardium, called M cells (654), are Potentials
variable in this respect, but they are still characterized
by a distinct phase 1 repolarization and a relatively large Arrhythmia research is often performed in different ani-
Ito (654), INaLate (658), and NCX (659) and small IKs (660). mal models, but its ultimate goal is to understand the
All these ion current characteristics contribute to the lon- mechanisms in humans and to prevent, or successfully
ger APD of M cells compared with subepicardial and treat, arrhythmias in patients. Therefore, it should always
subendocardial myocytes leading to a substantial trans- be kept in mind how experimental results can be extrap-
mural dispersion of repolarization (656). Augmented olated to humans. It is often overlooked that rodents
transmural dispersion of repolarization has been (rats and mice) have ion channel expression profiles

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1115


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

distinctly different from humans (40, 672). This also heart valve repair or replacement); therefore, human atrial
results in different cardiac electrophysiology properties cellular electrophysiological data are abundant, some-
(672, 673), especially during repolarization (FIGURE 12). what limiting the need for such studies in experimental
Mice and rats have a high heart rate (600 and 400 animals.
beats/min, corresponding to cycle lengths of 100 and Although the species differences in the shape of ven-
150 ms), 10-fold faster than in humans. As a conse- tricular action potentials are most striking between small
quence, mice and rats have very short ventricular and rodents and humans, important species differences also
atrial action potentials, to provide enough time for dias- exist in the action potentials between humans and other
tole. These short action potentials result from the pres- mammals. Guinea pig ventricular muscle, unlike human,
ence of transmembrane currents like Ito and IKur (144). In does not express Ito and does not exhibit a prominent
the ventricles of larger mammals (guinea pig, rabbit, phase 1 repolarization (144, 145); however, it expresses
dog, etc.) or humans, these channels are expressed less large IKs with distinct gating properties compared with
or not at all and also have a different molecular back- human (3, 214, 249). Rabbit Ito, unlike human, is con-
ground and functional role. Action potentials in mice or ducted mainly by Kv1.4 channels, and as a consequence
rats lack a plateau phase (144); therefore IKr and IKs are cycle length-dependent APD is markedly different from
not likely to operate despite the fact that expression of that observed in human ventricle (682). Pig ventricular
mRNA for these channels has been reported (674). muscle exhibits Ca21-activated Ito chloride current that
Notably, both IKr and IKs were observed in neonatal shapes phase 1 repolarization (683) but lacks 4-AP-sen-
mouse ventricle, but after further development neither sitive Ito despite abundant expression of Kv4.2 and
IKr nor IKs was detected in adult mouse ventricular myo- KChIP2 mRNA and proteins (684). The dog ventricular
cytes (675). Also, inward currents like ICa and INa have dif- muscle was found to express a considerably higher den-
ferent impact on ventricular repolarization than in other sity of IK1 compared with human (685). This results in a
mammals. Consequently, drugs can have marked spe- stronger repolarization reserve and consequently less
cies-dependent effects on action potentials as demon- APD prolongation upon IKr inhibition in the dog ventricle
strated with an example of IKr inhibition in FIGURE 13. compared to human. All of these differences have a par-
These fundamental differences mean that mice and rats ticular significance when drug effects and pathophysio-
can only be properly used in arrhythmia and related phar- logical electrophysiological alterations are extrapolated
macological research if the limitations of the models are from animal models to humans.
described. As FIGURE 13 illustrates, pharmacological in- Human induced pluripotent stem cell-derived cardiac
hibition of specific potassium channels, such as IKr, Ito, myocytes (HiPSCs) are increasingly used in cellular ar-
and IKur, elicits strikingly different effects on ventricular rhythmia research (686, 687). This new approach is
repolarization in the rat, a commonly used laboratory ex- promising and expanding rapidly (688). At the present
perimental animal, compared with humans. Despite this, stage, however, it seems that HiPSCs have some impor-
hundreds or thousands of papers using mice or rats have tant limitations (689). Although in HiPSCs experiments
been published on this topic, because these animals are can be performed relatively fast, at present they cannot
relatively cheap and easy to house. In addition, trans- provide a substitute for carefully applied animal prepara-
genic manipulations of transmembrane ion channels are tions. The so-far unresolved problems with HiPSCs are
almost entirely applied in mice (674), with very few excep- the following: data include cardiomyocytes that have not
tions (676–678). This makes the mouse a favorable tar- fully differentiated, still showing an immature phenotype
get, despite the fact that the mouse can be useful to (690, 691), and the cells spontaneously beat and often
study sodium and calcium channels and connexins but have a relatively low resting potential because they lack
not potassium channels. IK1 and also have low upstroke velocity (692–694).
There are far less consistent data on characteristic However, an interesting study suggests that the low
species-dependent differences in atrial action poten- resting potential and reduced IK1 are not necessarily in-
tials and the underlying specific transmembrane cur- herent characteristics of HiPSC-derived cardiomyocytes;
rents. As FIGURE 10 shows and several papers rather, these observations might be due to technical
indicate (297, 645, 679, 680), most of the species issues related to performing patch clamp on the rela-
commonly used in experimental laboratories exhibit tively smaller cells (692). Also, HiPSC sarcomeres are
similar action potentials and underlying currents, with disorganized, and their shapes are different from those
the atrial action potentials lacking a plateau phase, of the adult cells (691). So far, atrial and ventricular-like
except in humans (556, 681). Human atrial tissue sam- HiPSC cells have been successfully generated, whereas
ples, unlike ventricular samples, can be obtained from SAN, AV node, or Purkinje-like stem cell generation has
cardiac surgery departments (from patients undergoing been unsuccessful (691). However, HiPSC-derived myo-
open heart surgery for coronary artery bypass grafting, cytes from patients with defined mutations using

1116 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

CRISPR/Cas9-edited cells can mimic diseases (688, 5. COMPUTER SIMULATIONS OF ACTION


695–698) that are often hard or impossible to recreate POTENTIAL
properly in animal experiments. Recent efforts to culture
and continuously pace HiPSC-derived cardiomyocytes The vast quantity of experimental data describing struc-
cultured in collagen gels as “engineered heart tissue” ture and function of myocytes has enabled the develop-
represent a new two-dimensional (2-D) and three- ment of computer electrophysiology models capable of
dimensional (3-D) approach (699–701), since it resem- replicating action potential and conduction properties in
bles more the mature myocardium. Future research in a variety of species including human. Thus, the last 60 yr
this area, however, may revolutionize the field, opening have seen huge progress in our ability to model and sim-
new horizons for arrhythmia research. ulate the electrophysiology of the heart. Cellular

INa INaK INaCa IKs


IKb

I(Ca)Cl
INaL INab IK1 IKr
Ito
INaCa
Jrel IpCa
Jtr
IClb
ICaL

Jup ICab
Jleak

Jdiff ICaL

50 x 10-4
4
Membrane potential (mV)

3
0
Cai (mM)

-50

-100 0
0 100 200 300 400 500 0 100 200 300 400 500
Time (ms) Time (ms)

FIGURE 16. Structure of the Tomek, Rodriguez—following O’Hara–Rudy dynamic (ToR-ORd) ventricular myocyte model. The cell consists of 4 compart-
ments: main cytosolic pool of ions, SS [junctional subspace along T tubules, where Ca21 influx via L-type Ca21 current (ICa,L) and Ca21 release from the
sarcoplasmic reticulum (SR) interact, and where local calcium concentrations may reach much higher values than in the main cytosolic compartment], and
2 subcompartments of the SR [network (NSR) and junctional (JSR)]. I, ionic currents; J, ionic fluxes. Clouds correspond to calcium buffers. Key effects of
Ca21-calmodulin kinase-II on ionic currents and fluxes (such as ICa,L or SR release and reuptake) are also represented (717). Below the model diagram the
simulated membrane potential and calcium transient are shown, in agreement with experimental data. RyR, ryanodine receptor; SERCA, sarcoplasmic
reticulum CaATPase; CSQN, calsequestrin; ICab, background calcium current; I(Ca)Cl, calcium-sensitive chloride current; IClb, background chloride current;
IK1, inward rectifier potassium current; IKb, background potassium current; IKr, rapid delayed-rectifying potassium current; IKs, slow delayed-rectifying potas-
sium current; INa, sodium current; INab, background sodium current; INaK, sodium-potassium pump current; INaL, late sodium current; INaCa, sodium-calcium
exchange current; Ito, transient outward potassium current. Reproduced from Ref. 717 with permission.

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1117


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

computational models of different species and cell types clamp, or sine-wave protocols are also used to construct
are now available, through data and knowledge integra- models with complex gating properties (718, 719).
tion in an iterative process between experimental and Similarly to the necessity of collecting more complex
computational work (702). The first models of a cardiac data, a mathematical model of a current sometimes
cell were the Noble model of the Purkinje cell (703) and requires more complex structure. There are data that
the Beeler and Reuter model of ventricular muscle fiber cannot be represented accurately with the Hodgkin–
(704), following the pioneering work of Hodgkin and Huxley equations, and a more sophisticated modeling
Huxley in giant squid axon (705). Since those times, method is needed. In such cases, Markov models are
computer models have developed tremendously in their typically used instead, where distinct states of a channel
capabilities, robustness, and applications, ranging from are represented (e.g., open, closed, inactivated by volt-
understanding of arrhythmogenesis (706–712) to drug age, inactivated by drug, etc.) along with probabilities of
safety testing in industrial and regulatory settings (713– transitions between these states (719–721). Recently,
715). electrophysiological measurements were combined
Simulations with computational models offer perfect with molecular dynamics simulation (<0.25 Å) of the
observability and controllability, which help to overcome channel behavior to simulate ionic currents, which may
limitations in experimental models. They can be viewed allow simulations of mutations and drug effects repre-
as an organized and formalized literature review that senting the actual molecular processes (722, 723).
can be probed through simulation studies. They provide
mechanistically informed and tractable predictions, 5.1. Ventricular Models
which can then inform additional experiments and be
linked back to existing knowledge of physiological Given the relevance of ventricular fibrillation, the field of
mechanisms. At the same time, when simulations with a computational cardiac electrophysiology has focused
computational model fail to reproduce a particular phe- predominantly on ventricular cardiomyocytes. A mile-
nomenon, gaps in knowledge or inconsistencies in the stone in the development of ventricular models was the
state of the art can be identified. This may be an oppor- Luo–Rudy model of a guinea pig cell (707, 708), setting
tunity for discovery of key factors or processes not standards in how models are constructed and evaluated
included in the model, which may be critical for specific based on experimental data and how such models are
phenomena in living cells (716). used to study physiological behaviors. These models
A computational model of cardiac cellular electro- include the key ionic currents, such as fast sodium cur-
physiology typically consists of a set of differential rent, L-type calcium current, delayed repolarization cur-
equations describing the mechanisms of transmem- rent, sodium-calcium exchanger, and the inward rectifier
brane ionic transport and excitation-contraction cou- potassium current. The main features of calcium handling,
pling. Today’s models are commonly separated into including sarcoplasmic reticulum release and reuptake,
multiple subcellular compartments to allow local- are also incorporated. The model was subsequently used
ized calcium signaling and diffusion {an example of to study the interplay of ionic currents and ion concentra-
the structure of the recent human ventricular myo- tions in formation of early and delayed afterdepolariza-
cyte model [Tomek, Rodriguez—following O’Hara– tions (707, 708). The Hund–Rudy canine model (724)
Rudy dynamic (ORd) (ToR-ORd)] (717) is reproduced separated the delayed rectifier currents into rapid (IKr)
in FIGURE 16}. and slow (IKs) components (as in Ref. 725), added two
The most broadly used framework of modeling dis- components of transient outward current (Ito,1 and Ito,2),
tinct ionic currents is the Hodgkin–Huxley equations and included a model of CaMKII activation and signaling,
(705), where several independent “gates” (activation, studying the impact on rate-dependent behaviors. In
inactivation, etc.) of the respective current are formu- addition, it introduced an intracellular subcompartment:
lated based on experimental data; a combination of the the junctional subspace (where L-type calcium channels
gates then determines the fraction of open channels. and ryanodine receptors colocalize and where calcium
This is subsequently combined with open channel con- concentration reaches a much higher value than in the
ductance and ionic driving force through the open chan- bulk cytosol), which is also a standard in today’s models.
nel to produce total current. Traditionally, single-pulse The model was subsequently updated (726) and used to
square voltage clamp was used to collect data used to study the effect of disease (727) and of b-adrenergic stim-
create ionic current models. However, in some cases it ulation (728).
was shown that predictions based on such data do not Another very influential model, created in 2004, is the
match behavior of the current under AP clamp, such as Shannon rabbit model (729), which also separated differ-
in the late sodium current (693). Consequently, more ent components of delayed rectifier current and
complex protocols such as two-pulse voltage clamp, AP included the junctional subspace and which focused

1118 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

predominantly on an accurate representation of calcium 5.3. Other Tissue-Specific Modeling


handling. The model was subsequently modified to
improve its rate-dependent behavior (721) and was used As discussed in sect. 4 of this review, different tissues
to study alternans. Interestingly, even though a large manifest markedly different action potentials, achieved
proportion of experimental cardiac research is carried by differential function of ionic currents. Cardiac models
out in rodents, there are very few detailed rodent mod- reflect this, allowing tissue-specific simulations repre-
els. Relatively recent models by Bondarenko et al. (730) senting specific features of atrial, Purkinje, or sinoatrial
and Morotti et al. (731) thus provide an important tool in cells.
complementing rodent-based experimental research,
and they may be used to gain insight into species differ- 5.3.1. Atrial models.
ences (40).
Human atrial electrophysiology modeling and simula-
5.2. Human-Specific Ventricular Models tions are active topics of research. Two prominent mod-
els of human atrial electrophysiology are by
Modeling and simulation of human ventricular electro- Courtemanche et al. (738) and Nygren at al. (739), both
physiology are crucial subcategories of computational recapitulating specific AP morphology of atrial myo-
cardiac electrophysiology. The central species difference cytes. The Courtemanche model used the Luo–Rudy94
between human and other mammals is the high reliance model as a starting point (708) but reformulated the ionic
of human cardiomyocytes on IKr for repolarization, unlike currents according to human and canine data from atrial
in other species, where the also relatively highly myocytes. The model was used to understand the im-
expressed IKs provides an additional repolarization portance of L-type current inactivation in APD shorten-
reserve. This is of utmost importance, e.g., in the study of ing in the S1S2 protocol and to gain insight into the role
drug safety, where hERG/IKr blockers may cause a much of Ito in variability of atrial AP morphology. The Nygren
greater APD prolongation in humans than in animals model (739) was mostly based on a similar set of data,
(685). The specifics of human repolarization thus warrant but it used a different model as the starting point (740).
the development of specifically human-driven computer Its analysis provided insight into differences between
models. The first of these that gained wide popularity human and rabbit repolarization, focusing on the role of
was the pair of models by ten Tusscher et al. (732, 733). A the sustained outward potassium current. More recent
second family of widely used human ventricular myocytes models, adding and studying ionic homeostasis, addi-
is the model by Grandi et al. (734) and the Carro– tional currents, and signaling, are described in Refs.
Rodríguez–Laguna–Pueyo (CRLP) model that followed it, 741–743. We refer the reader to more comprehensive
focusing on rate dependence and behavior in hyperkale- reviews (744–746) on atrial modeling and simulation.
mic conditions (735). Possibly the currently most popular
human ventricular model is the one by O’Hara et al. (ORd) 5.3.2. Purkinje cell models.
(736), created and evaluated with a wide range of experi-
mental data, many of which were collected specifically for Even though Purkinje fiber cells were the focus of the
the purpose of the model development. The ORd model first cardiac electrophysiology models (703), progress
incorporates CaMKII signaling and is capable of manifest- in this area has been irregular. In 1985 an updated
ing early afterdepolarizations among other features. model was published, describing both electrophysiol-
Together with its humanlike mixture of repolarization cur- ogy and changes in ionic concentrations (639), but the
rents, it has become, with various modifications, a domi- focus subsequently shifted toward other cell types
nant model in the assessment of drug safety (713, 715) discussed above (747). In recent years, the interest in
and is a prime example of how computer modeling and Purkinje fibers has been reinvigorated, producing
simulation may be used for regulatory purposes (737). models focusing on their role and changes in heart
Recently, a new human ventricular myocyte model, ToR- failure (748), the electrophysiological basis of rabbit
ORd, was published (717), improving on its predecessor Purkinje electrophysiology (749), and the importance
ORd in multiple aspects, such as similarity of the action of Purkinje fibers in arrhythmogenesis (750, 751). The
potential morphology to experimental data or the very recent Purkinje model by Trovato et al. (752)
response to sodium channel blockers. The model also introduced a comprehensive calibration to human
brought biophysical insight into properties of ICa,L and IKr, data in a wide range of conditions, bringing insights
which are important for the understanding of sodium and into Purkinje cell EADs and abnormal automaticity
calcium dynamics in ventricular myocytes. such as following IK1 reduction or If increase.

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1119


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

5.3.3. Sinoatrial cell models. are multiple sources of variability in experimental and clin-
ical data, caused by both intra- and interheart differences
Sinoatrial cell modeling and simulation has also made sig- as well as differences in experimental settings and/or
nificant progress in recent years in both rabbit (753, 754) measurement techniques (50). One of the techniques to
and human (755), enabling investigations into the ionic ba- investigate potential causes and modulators of biological
sis of the cardiac pacemaker in normal and disease condi- variability is the use of populations of models rather than
tions including the effect of mutations (281, 755, 756). a single generic computational model (768, 774, 775).
Populations of models are ensembles of models sharing
5.3.4. HiPSC models. the same structure based on a particular cardiomyocyte
model (such as the O’Hara–Rudy model) but with varia-
An additional cell type that is gaining increasing popular- tions in model parameters, such as the conductances of
ity is human-induced pluripotent stem cells (HiPSCs), ionic currents. To achieve plausible populations, a calibra-
which offer a new human-based paradigm in experimen- tion to experimental or clinical data can be carried out
tal research. Computer modeling and simulation of (768): e.g., a model is accepted in a population only if its
HiPSC is an important tool complementing the experi- action potential morphology and calcium transient prop-
mental system. This was pioneered by Paci et al. (757) erties fall within an experimentally observed range. Such
and is now an active research field (758, 759). calibrated populations have proven to be useful in the
study of hypertrophic cardiomyopathy (776) and atrial fi-
brillation (775, 777) and for drug safety assessment (715,
5.4. Different Modeling Scales 775). An intrinsic limitation is that a plausible phenotype
may be produced by an unplausible mechanism and/or
In sect. 5.3, we focused predominantly on cell-level by a biologically unrealistic combination of conductances
modeling, where one set of equations describes the of ionic currents. More comprehensive data collection,
cellwide current. This usually provides a good trade-off reporting, and understanding of biological variability is
of relatively high biological detail but still affordable thus an important future step for the fields of both experi-
computational time (<1 s per 1 action potential). For spe- mental and computational cardiac electrophysiology.
cific purposes, it may be necessary, however, to either
increase or decrease the level of detail. In particular,
detailed phenomena driven by cellular calcium handling 6. CELLULAR ARRHYTHMIA MECHANISMS
(such as calcium waves and resulting delayed afterde-
polarization as well as some mechanisms of calcium-
6.1. Depolarization Abnormalities
driven alternans) usually require a high degree of detail
(760–762). In such models, a single cell is subdivided
Impaired depolarization capability in cardiomyocytes can
into small (microscale or even nanoscale) cuboids that
reduce conduction safety, providing proarrhythmic sub-
may represent membrane, calcium release units from
strate and potentially contributing to conduction block and
the SR, cytosol, etc. and are coupled together to repre-
reentry arrhythmia. One example of depolarization abnor-
sent the cell. Such a level of detail, however, consider-
mality leading to reentry arrhythmia is the Brugada syn-
ably increases computational time, such as hours
drome, caused by the loss of function of SCN5A (778,
(microscale models) or more than a day (nanoscale) for
779). It should be noted, however, that in patients with
several beats on a personal computer. On the other
Brugada syndrome the mechanisms of arrhythmia are
side of the spectrum of model complexity are models
more complex, since decreased function of L-type ICa and
such as the Fenton–Karma (763) and minimal ventricu-
enhanced function of Ito also contribute to the develop-
lar (MV) (764) models, which are more than an order of
ment of phase 2 reentry (780), due to enhancement of dis-
magnitude faster to simulate compared with biologically
persion of repolarization across the ventricular wall (779).
detailed cellular level models. This makes such models
Ischemia can cause regional membrane depolariza-
an attractive tool, for example, model personalization
tions, which indirectly decrease the strength of INa by
using clinical electrophysiological recordings (765) or
eliciting partial or full channel inactivation and result in
when tissue properties (rather than ionic properties) are
slowing of impulse conduction or unidirectional or bidir-
the target of the investigations (766, 767).
ectional conduction block. All of these factors are con-
sidered important in arrhythmogenesis. However, their
5.5. Variability in Computer Models detailed discussion is beyond the scope of this review,
since they are described by others in great detail (31,
In recent years, representing variability in cardiac models 781–785). Drugs that inhibit INa can have similar effects
has emerged as a key new concept (50, 768–773). There and can also cause reentry arrhythmias (202, 786, 787).

1120 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

Upregulation of the If current (788) and HCN channels example, Kurata et al. (797) demonstrated how NCX con-
(789) in the ventricles and atria (790) was reported in HF tributes to EADs in the popular O’Hara–Rudy model (736)
(789) and HCM (791, 792). These changes can cause via two distinct mechanisms. First, the influx of calcium via
abnormal myocardial depolarizations, and they can the L-type calcium current upon its reactivation translates
relate to increased incidence of ectopic beats, providing into calcium efflux via NCX and thus additional inward
possible triggers for arrhythmias in an environment current (which can, in turn, promote further activation of L-
where dispersion of repolarization (arrhythmia substrate) type calcium current). Second, NCX expressed adjacent
is already augmented by structural heart disease. to L-type calcium channels acts as a “sanitizer” of calcium
in the cellular region, removing calcium ions from the
6.2. Triggered Automaticity junctional subspace during repolarization. This may sub-
sequently reduce the calcium-dependent inactivation of
Abnormal myocardial automaticity (formation of prop- L-type calcium current, facilitating earlier reactivation. The
agating spontaneous action potential) is an estab- insight into EAD origins is not limited to L-type calcium
lished trigger contributing to arrhythmia onset (26). current and NCX; nonequilibrium gating of late sodium
One particular type of such automaticity closely linked current was implicated in EAD formations (798). The
to disturbance of normal cellular electrophysiology is increased availability of data on signaling pathways has
the so-called “triggered automaticity,” which requires also enabled computationally driven insights on how
preceding action potentials that are essential for the EADs are facilitated via CaMKII- or PKA-driven pathways
subsequent spontaneous firing (793). Depending on (799). This is particularly important for our understanding
the temporal relationship between such depolariza- of EADs in disease conditions such as heart failure, where
tion and the preceding action potential, triggered au- these pathways are dysregulated.
tomaticity is typically separated into early and delayed One interesting involvement of IKr in EAD formation
afterdepolarizations. beyond the role in APD prolongation lies in its dynamic
of activation and reactivation. Lu et al. (719) used a range
6.2.1. Early afterdepolarizations. of electrophysiological protocols to demonstrate an in-
triguing interplay of IKr activation, inactivation, and recov-
Early afterdepolarization (EAD; FIGURE 10, A and B) is ery from inactivation, which leads to rapid increase of IKr
characterized by depolarizing potential changes occur- during late-plateau reactivation, such as during an EAD.
ring before the termination of the preceding action Such an increase in repolarizing current would be
potential during phase 2 or phase 3 repolarization. expected to counteract and potentially outweigh depo-
EADs are usually generated when action potential dura- larizing currents, potentially preventing the formation of
tion is excessively prolonged, e.g., when IKr is impaired. a larger-amplitude EAD.
As a consequence of the lengthened action potential,
those L-type calcium channels that have already recov- 6.2.2. Delayed afterdepolarizations.
ered from inactivation can reopen, and some of the cal-
cium channels carry a Ca21 window current (707, 708), The delayed afterdepolarization (DAD) is characterized
causing positive voltage oscillations during the plateau by depolarizing potential changes following the termina-
(phase 2 EAD) or terminal repolarization (phase 3 EAD). tion of the preceding action potential (129, 130, 632) dur-
The Luo–Rudy studies (707, 708) also proposed a sec- ing diastole (FIGURE 10C). DAD is generally attributed
ond type of EADs, phase 3, resulting from spontaneous to calcium-sensitive depolarizing currents after sponta-
calcium release during repolarization, which then trans- neous Ca21 release from the SR during Ca21 overload,
lates into depolarization via NCX. The relevance of this or CaMKII-dependent phosphorylation (800, 801), which
mechanism was subsequently demonstrated experi- is promoted by diseases like chronic AF, ischemia, heart
mentally in Purkinje fibers (131). This type of EAD strongly failure (801–803), and catecholaminergic polymorphic
resembles delayed afterdepolarizations in its mecha- ventricular tachycardia (CPVT) or drugs like digitalis. The
nism but differs in the timing (during AP vs. after AP). most important calcium-sensitive current implicated in
Both types of EADs are rate dependent, with the reacti- DAD formation is the forward-mode NCX, but a role for
vation-driven mechanism appearing predominantly at the calcium-sensitive chloride current has also been
slow pacing, whereas the release-driven EADs occur at suggested (804). The calcium-induced depolarization is
fast pacing (794). opposed primarily by IK1, which tries to maintain resting
In general, both the L-type calcium current and NCX potential (805). When the depolarization induced by cal-
are known to act synergistically in EAD formation (795, cium-sensitive currents is of sufficient magnitude, over-
796), and the controllability of computational models coming IK1, it activates INa, triggering a new action
has been used to understand their interplay. For potential. Automaticity occurring in the pulmonary veins

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1121


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

and in the myocytes represents important abnormal investigated the effect of gap junction uncoupling, fibro-
impulse formations, and at present it seems that their sis, and heart failure-like remodeling, showing that the
cellular mechanisms are complex, including the possibil- combined effect may reduce the number of cells
ity of DAD and EAD generation as well (FIGURE 14). needed for an afterdepolarization-driven propagation by
The spontaneous calcium release is a stochastic phe- two orders of magnitude. Specific patterns of uncou-
nomenon, represented by calcium sparks (806, 807) pling, such as thin strands of myocytes within postinfarc-
and intracellular calcium waves (807–810), and multiple tion scars (817), which are similar to a fiber with regard to
calcium release sites need to synchronize to produce a coupling, may increase the relevance of afterdepolariza-
cellwide calcium release (811, 812). The stochastic nature tions of arrhythmia even further. Another type of weakly
of such events as well as generally limited controllability coupled tissue that might enable synchronization of
and observability of subcellular calcium handling in the afterdepolarizations is the endocardial Purkinje ventricu-
experimental setting, complicate detailed understanding lar junctions, as mentioned above (FIGURE 15).
of their origins. On the other hand, computer models In addition to promotion of extrasystolic reactivation
(736) offer excellent controllability and observability and via cell decoupling, other mechanisms of synchroniza-
thus are a popular tool to understand origins of sponta- tion of afterdepolarizations are via stretch-activated
neous calcium release and ultimately DADs. Increasing channels (818), current flow in the border zone of acute
availability of subcellular experimental data has enabled ischemia (706), and partial chaos synchronization (369).
the construction of spatially detailed models, where the In a modeling study of calcium-driven afterdepolariza-
cell is subdivided into up to hundreds of thousands of tions, it was suggested that calcium waves also syn-
subdomains with separate clusters of potentially sto- chronize by calcium flux through gap junctions (819);
chastic ryanodine receptors (760, 813, 814). As a result, however, experimental results argued against this possi-
such models can give very detailed predictions, eluci- bility (811). Ultimately, simulation studies have shown that
dating how originally random calcium sparks are the baseline risk of EADs may be considerably incr-
recruited into calcium waves and ultimately DADs but eased in diseased conditions (776, 820), further facilitat-
also giving explanations for DADs that do not rely on cal- ing translation of a depolarization to tissue activation.
cium waves (814). In the case of increase in intracellular calcium during
elevated sympathetic drive and/or diseases like heart
6.2.3. From afterdepolarizations to arrhythmic failure or catecholaminergic polymorphic ventricular
behavior. tachycardia (CPVT) (821), Ca21 overload may occur and
the SR can become leaky and release additional Ca21
One important aspect of triggered automaticity is that (822). Mutations in the gene encoding the cardiac ryano-
the role of EAD and DADs in arrhythmogenesis is most dine receptor-related Ca21 release channels (RyRs) can
likely overestimated in single-cell experiments versus cause extrasystole and serious tachycardia such as
the intact heart. Even in relatively poorly coupled tissue, CPVT by abnormally releasing Ca21 from the SR (823)
electrotonic interactions with neighboring cells will into the cytosol on the response of catecholamines
decrease the depolarization produced by an EAD or a which Ca21 would activate the electrogenic forward
DAD. However, moderate uncoupling will decrease the NCX depolarizing cells beyond their threshold of activa-
electrotonic interaction between a focus and the sur- tion. In a recent study, in a new model for CPVT in engi-
rounding cells and can actually favor action potential neered human tissue fabricated from human pluripotent
propagation (815). stem cell-derived cardiomyocytes, high-frequency pac-
Computer simulations studies have significantly con- ing and isoproterenol administration increased Ca21
tributed to the understanding of the conditions under wave propagation heterogeneity and elevated intracel-
which the afterdepolarizations of single cells may trans- lular [Ca21], leading to local depolarizations and conduc-
late into propagation throughout myocardium (816). tion block (creating the arrhythmia substrate), and
These modeling studies showed that when simulating subsequently resulting in reentry (700). Similarly to
healthy cells and their afterdepolarizations, 70 cells CPVT, leaky ryanodine receptor-related Ca21 release
manifesting an afterdepolarization are needed to trigger channels were also reported in heart failure (802, 824).
excitation in a fiber, 7,000 in 2-D tissue, and
700,000 in 3-D tissue. The specific numbers may very 6.3. Frequency Dependence and Restitution
well vary with numerical aspects related to the simula-
tions, such as the mesh discretization, but this neverthe- It was observed long ago that action potential duration
less suggests the unlikeliness of EADs or DADs and impulse conduction depend on heart rate or on the
promoting into tissue reactivation in a healthy tissue. At stimulation frequency. To study frequency-independent
the same time, however, the study by Xie et al. (816) also repolarization changes caused by disease, drugs, or any

1122 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

Frequency dependent APD Abrupt change of cycle length


(electrical restitution)
320 300

280 FIGURE 17. Cycle length-dependent action


potential duration (APD90) changes (left) in
280 undiseased human (donor) ventricular mus-
260
cle (open symbols) and Purkinje fiber (filled
symbols) preparations. Right: the relation-
240 ship between APD90 and the preceding dia-
APD90 (ms)

APD90 (ms)
stolic interval (S1S2 restitution) in human
240 ventricle (open symbols) and Purkinje fibers
220
(filled symbols). Both protocols were meas-
ured with the conventional microelectrode
200 technique. APD90, APD at 90% repolariza-
tion. Unpublished observations from our lab-
200
Basic cycle length: 1000 ms oratory at the Department of Pharmacology
180
and Pharmacotherapy, University of Szeged,
Human Purkinje Human Purkinje Hungary.
Human ventricle Human ventricle
160
160
0 1000 2000 3000 4000 5000 0 40 80 120 160 200

Cycle length (ms) Diastolic interval (ms)

other factors, correction formulas have been used to esti- decreased safety or slowed impulse propagation.
mate QT intervals corrected for heart rate (QTc) on the Frequency-dependent APD changes show general pat-
ECG. At elevated heart rate, extracellular K1 accumula- terns (682, 825–830) that APD is short at high and longer
tion may occur in the clefts, slightly depolarizing the rest- at slow constant rates (FIGURE 17). The frequency de-
ing membrane potential that slows impulse conduction pendence of APD shows substantial species, tissue, and
and impairs safety of impulse propagation. At extrasys- regional variation and has important implication for
toles early following the end of ERP, repolarization is still arrhythmogenesis. At slower heart rate, APD can be
not fully terminated and Na1 and Ca21 channels are par- markedly prolonged, favoring triggered arrhythmias via
tially inactivated, resulting in less depolarizing current and EAD formation, and may also result in enhanced

R1: Steep restitution curve R2: Flat restitution curve


FIGURE 18. Arrhythmia development
APD (ms)

APD (ms)

via regional differences of the action


potential (AP) restitution. Left: consecu-
tive action potentials from a region with
a steep restitution curve (R ). Right: a
DI (ms) DI (ms) 1
region with flat action potential duration
S1 S2 S3 E1 E2 S1 S2 S3 E1 E2 (APD) restitution (R2). In both cases, S1–
S indicate 3 consecutive sinus node
3
stimuli followed by 2 extra stimuli (E1
and E2). In the case of steep APD resti-
tution, the E is able to propagate while
1
the conduction of E2 is blocked since
the refractory period of the developed
R1 R2
extra AP is sufficiently long. In contrast,
in the flat restitution region both extra
stimuli will propagate, since the APD of
the extra beats are not markedly pro-
longed. Therefore, these APs can reach
the R region (red arrow) and evoke
1
extra beats where the refractory period
has already been terminated (red
curves), establishing a circulating move-
ment of impulse propagation. DI, dia-
stolic interval.

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1123


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

FIGURE 19. Proposed mechanism of action potential (AP) alternans and arrhythmia development. The top sequence of the AP illustrates a proximal
and the bottom sequence a distal area of the left ventricle in respect of impulse propagation. On left, 2 consecutive normal (N) action potentials are
shown; the depolarization is readily conducted toward the distal area (black arrows), evoking further APs. When an extra stimulus (ES) with short cou-
pling interval reaches the proximal area, the evoked APs will be organized as a short-long-short pattern in both regions, establishing concordant alter-
nans (green curves). Under this condition, a second extra beat will evoke a short AP in the proximal region but a long AP in the distal area, since the
refractory period causes slowed impulse propagation (discordant alternans, orange curves). In this case the heterogeneity of the repolarization causes
a third extrasystole (red curve) in the proximal region to be blocked in the distal area (red dashed arrow), providing possibility for development of reen-
try arrhythmia. APD, action potential duration.

substrate for arrhythmias by increasing dispersion of rep- is considered proarrhythmic (20, 662, 837, 838), whereas
olarization. flattened electrical restitution curves would have an op-
Electrical restitution refers to the recovery of the APD posite consequence (FIGURE 18). Local regional differen-
of an interpolated beat as a function of time following the ces in the APD restitution curves (839) may also favor
previous beat. This changes in a manner that is some- arrhythmogenesis (840). The ion channel background of
what similar to that seen (831) during frequency-depend- cycle length-dependent APD changes including APD res-
ent steady-state APD changes (FIGURE 17, left). Despite titution is attributed to the incomplete recovery and/or
the similarities, there are important differences (FIGURE deactivation of different inward (INa, ICa,L) or outward (Ito,
17, right) that warrant the restitution being studied as a IKr, IKs, ICl) currents based on the gating behavior of these
separate rate-dependent property (826, 827, 832–835), channels (841, 842). In addition, intracellular ion concen-
with importance for arrhythmia research (20, 836). tration changes for Ca21 and Na1 rapidly or slowly would
According to the restitution hypothesis, as the diastolic activate electrogenic NCX or Na1-K1 pumps. Also, fre-
interval increases because of the propagation of an extra quency changes can result in significant alterations in
beat, the second extrasystole would encounter longer extracellular K1 concentration in the extra-cellular clefts
APD/ERP and local conduction block may occur. A (843), causing changes not only in depolarizing but also
steeper restitution curve would favor such an effect and in repolarizing transmembrane ionic currents. Detailed

1124 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

discussion of these mechanisms is beyond the scope of suggested that refractoriness of the ryanodine receptor
this review. rather than release-reuptake mismatch may underlie at
least some of the observed alternans. Such a mecha-
6.4. Repolarization Alternans and Temporal nism is also supported by certain computer models and
Repolarization Variability may be either due to the refractoriness of the channel or
due to changes in calsequestrin conformations with sub-
Repolarization alternans (FIGURE 19) at the cellular level sequent RyR block from within the SR (761). In a recent
manifests as oscillation of long and short APD at rapid study utilizing iterated maps as well as a spatially
heart rates, typically with concurrent oscillations in cal- detailed myocyte model, Qu et al. (856) showed that
cium transient amplitude (712, 844). It has been demon- both mechanisms may act synergistically to promote
strated that alternans can precede the formation of alternans. A third explanation of calcium-driven alter-
arrhythmia in the heart (845, 846). Multiple studies and nans is the alternans driven by sarcoplasmic reticulum
reviews explain the mechanisms of arrhythmia induction calcium cycling refractoriness (SRCCR) (857, 858).
following alternans, typically linked to increased disper- SRCCR alternans arises from a combination of steep
sion of repolarization (712, 847, 848). The spatial pattern load-release relationship (similar to release-reuptake
of alternans across cardiac tissue is typically “concord- mismatch hypothesis) and refractoriness of the SR
ant” at submaximal heart rates, i.e., the APD is either release (similar to RyR refractoriness hypothesis).
simultaneously shortened or prolonged in all sites However, the latter does not result from an intrinsic RyR
(FIGURE 19) (712). However, further increase in the pac- refractoriness but is a result of a limited rate of refilling
ing rate can elicit so-called discordant APD alternans of releasable calcium in the junctional SR. This mecha-
(FIGURE 19), when APDs at more distant regions can nism underlies alternans in the Rudy-family models (728,
alternate with opposing phases (712), substantially 736, 857).
increasing dispersion of repolarization and thereby the Alternans typically occurs only at rapid heart rates;
substrate for arrhythmias (849). however, data from human hearts show that in some
The fact that repolarization alternans typically occurs cases alternans manifesting at rapid heart rate may cease
together with underlying oscillation of calcium transient with a further increase in pacing frequency (“eye-type
amplitude (850, 851) poses the question of which of alternans”) (859). The eye-type pattern was replicated
these two drives the other. with a populations-of-models approach (859), with the
The first hypothesis suggests that the steep slope of mechanism of the eye closure at rapid pacing being
APD restitution is the alternans driver (852), and this linked to flattening of the SR load-release relationship in
mechanism of arrhythmia is replicated by the ten certain conditions (858). Using a spatially distributed
Tusscher–Noble–Noble–Panfilov model of human ven- model of calcium handling, Qu et al. (856) also observed
tricular myocyte (732, 733). As mentioned above, the ion eye-type alternans in simulations where sarcoplasmic
channel background of APD alternans is attributed to reticulum Ca21-ATPase (SERCA) pumps were
the incomplete recovery and/or deactivation of different downregulated.
inward (INa, ICa) or outward (Ito, IKr, IKs, ICl) currents based One important question pertaining to the spatial pat-
on the gating behavior of these channels (841, 842). In tern of alternans in tissue is what determines whether
addition, intracellular ion concentration changes for alternans manifests in the relatively benign spatially con-
Ca21 and Na1 rapidly or slowly would activate electro- cordant pattern or the highly proarrhythmic discordant
genic NCX or Na1-K1 pumps. Also, frequency changes one. Pastore et al. (849) observed in their experiments
can result in significant alterations in extracellular K1 that nodal lines (lines in tissue separating areas of
concentration in the extracellular clefts (843), causing opposing alternans phase) were associated with struc-
changes not only in depolarizing but also in repolarizing
tural abnormalities. However, discordant alternans may
transmembrane ionic currents.
arise even in tissue with no obvious structural abnormal-
On the other hand, other studies suggested that oscil-
ity. Computer models provide two explanations of this
lations in the calcium transient amplitude are the primary
phenomenon. Qu et al. (860) have shown that discord-
alternans driver (851, 853). Such oscillation of calcium
ant alternans may emerge as a result of conduction ve-
transient can be subsequently translated into APD alter-
locity restitution. This explanation is characterized by
nans by NCX and other calcium-sensitive currents.
the radial pattern of nodal lines with regard to pacing
Calcium-driven alternans was first suggested to origi-
site. The second explanation by Sato et al. (762) relies
nate from a Ca21 release-reuptake mismatch due to the
on tissue synchronization of discordant alternans arising
steep dependence of Ca21 release on SR loading (851,
at the subcellular level. This explanation does not rely
854). While in good agreement with a majority of experi-
on a particular pattern of nodal lines and can explain ex-
mental data, the experiments by Picht et al. (855)
perimental observations in Ref. 847.

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1125


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

FIGURE 20. Short-term temporal variability of repolarization and the risk of sudden cardiac death (SCD). Top left: schematic illustration of QT interval
variability on an electrocardiogram trace. Top right: schematic illustration of action potential duration (APD) variability. Bottom left: increased QT interval
short-term variability in a representative professional soccer player compared with a control individual, illustrated on a Poincare  plot, where each QT
interval n is plotted against its former value, n 1. Bottom right: individual Poincare  plots illustrating increased short-term variability of left ventricular
(LV) monophasic action potential duration (MAPD) in chronic atrioventricular block dogs with SCD compared with control dogs without SCD. (Modified
from Ref. 861 with permission.)

Spontaneous episodes of ventricular tachyarrhythmia densities or functions of transmembrane ion channels


in patients are often preceded by a short-long-short and/or pumps, so-called electrical remodeling (TABLE 2)
sequence of cardiac cycles or irregular beat-to-beat vari- (7). Thus, electrophysiological remodeling is the result of
ation of the QT intervals. This temporal instability of changes in the expression of ion channels and pumps,
repolarization measured as short-term APD or QT vari- and it can affect both repolarization and impulse con-
ability similarly to spatial repolarization inhomogeneity is duction. In parallel, remodeling often affects the struc-
considered as an important marker for proarrhythmia ture of cardiac tissue, which can be detected by
(250, 861) (FIGURE 20). The mechanisms of short-term microscopy (880). Structural remodeling includes fibro-
APD variability are not fully understood; however, they sis and wound healing, resulting mainly from the activity
may relate to the stochastic behavior of the ion channels of fibroblasts and myofibroblasts (681, 881, 882). Fibrosis
underlying the action potentials or the fluctuation of the and scars impair impulse conduction, eliciting conduc-
intracellular Ca21 movements and its electrophysiologi- tion blocks, changing the directions of normal impulse
cal consequences (822). propagation, and enhancing the heterogeneity of
impulse conduction. Finally, remodeling of the postin-
farction border zone includes alterations to the pattern
7. ION CHANNEL AND ACTION POTENTIAL and function of gap junctions, further increasing hetero-
REMODELING IN ACQUIRED CONDITIONS geneity of conduction (883). The combined effects of
electrical and structural remodeling increase the pro-
Certain diseases and alterations in physiological func- pensity of reentry-based arrhythmias.
tions evoke changes in the heart that are collectively In this section, electrical remodeling following from
termed remodeling (7). Remodeling can also affect the the best-explored and probably most important

1126 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

Table 2. Remodeling of transmembrane currents/transporters in disease


AF-Atrial Muscle HF-Ventricular Muscle DM-Ventricular Muscle
Current
Reference Reference Reference

INa ; 7 ; 7, 862 ; 863,864

INaL : 126,127

ICa,L ; 7 Ø Ø slow inactiv. 865

Ito ; 7, 549 ; 7, 156, 159, 866 ; 271, 867,868

IK1 : 205 ; 7, 157

IKr ; 7 ;Ø 271, 867, 869

IKs ; 7 ; 271, 867

IKur ;Ø 549, 870

IK,Ach : 358, 584, 871

IKCa (SK2) : 562, 872 : 315, 873 ; 874

If : 788,789, 875,876

NCX : 7, 365

Connexin ; 7 ; 877–879

AF, atrial fibrillation; DM, diabetes mellitus; HF, heart failure; ICa,L, L-type Ca21 current; If, funny/pacemaker current; IK,ACh, acetylcholine-activated potas-
sium current; IKCa, calcium-activated potassium current; IKr, rapid component of delayed rectifier potassium current; IKs, slow component of delayed recti-
fier potassium current; IKur, ultrarapid component of delayed rectifier potassium current; IK1, inward rectifier potassium current; INa, sodium current; INaL,
late sodium current; Ito, transient outward current; NCX, Na1/Ca21 exchanger.

diseases is briefly summarized in order to better under- dog hearts, atrial APD is abbreviated less (1135) than
stand the mechanisms of cardiac arrhythmias. those observed in human (7, 309). This emphasizes
the importance of the substantial species differences
7.1. Remodeling in Atrial Fibrillation mentioned above, in this case between dog and
human atrial action potential waveforms (FIGURE 12).
AF is the most common sustained arrhythmia in the Therefore, the interpretation of results based on ani-
developed world, and its prevalence increases with mal experiments should be considered with care in not
age, exceeding 10% after the age of 80 yr (6, 884). AF only pharmacological but pathophysiological studies
itself is seldom directly life-threatening and usually as well.
has few acute hemodynamic consequences, but it High atrial rate or tachypacing in experimental animals
represents a high risk for thromboembolism and induces atrial remodeling (7, 363, 777, 887, 888) that is
stroke given an increased risk of thrombus formation characteristic for chronic AF. Atrial extrasystoles or elec-
in the hypodynamic atria (885). It can furthermore con- trical stimulation can easily induce AF in remodeled
tribute to the development of heart failure; therefore atria, and the longer and more often AF episodes are
AF has great significance in cardiovascular morbidity present, the more pronounced atrial remodeling and vul-
and mortality (886). nerability for further AF development become (“AF
Studies performed in atrial tissue originating from begets AF”) (889).
chronic AF patients or experimental animals show sig- Remodeling in AF is a complex and still not fully eluci-
nificantly shorter and often triangular-shaped action dated process (7, 30, 34, 887, 890–892). The most well-
potentials compared with those obtained from sinus established phenomenon in AF is downregulation of L-
rate patients and animals. It must be emphasized that type ICa and the corresponding mRNA and proteins. This
data regarding electrophysiological remodeling can occurs irrespective of the underlying cause of AF and is
be controversial. For example, in pacing-induced AF in observed in AF both with and without heart failure. It is

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1127


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

considered to be the main cause of APD shortening in fibroblast/myofibroblast activity, fibrosis, and altered
chronic AF. Also, the upregulation of Kir2.1 (the main connexin expression (877, 879) impair impulse con-
channel protein of IK1) and TASK-1 channels has been duction (881) and contribute to impulse propagation het-
reported to contribute to atrial APD shortening in AF erogeneity (901, 902). These changes alone and in com-
(205). The overall IK,ACh channel (Kir3.1/3.4 1 GIRK) bination with heterogeneity of repolarization (25, 903)
expression is not significantly changed in chronic AF would result in a favorable substrate for reentry arrhyth-
(871), but a constitutively active component of this cur- mias (FIGURE 2). An additional source of proarrhythmic
rent that is very weak in sinus rhythm is greatly substrate is the high vulnerability of failing hearts to
enhanced (358) and is assumed to shorten the APD in repolarization alternans (846, 904), which appears to
AF. A very recent study also indicated that cholinergic result mainly from remodeling of calcium handling. For
M1 receptors were upregulated, further activating IK,ACh example, Nivala et al. (905) utilized a detailed model of
and thereby shortening atrial repolarization (871). Both spatiotemporal calcium handling to investigate the con-
downregulation (549, 870) and no change of IKur have sequences of heart failure remodeling. They have
been observed in chronic AF. A recent study in a tachy- shown complex interactions of disruption of T tubules
paced dog AF model (562) confirmed the expression together with SERCA downregulation to promote
changes of SK2 or SK3 channels, which supported the increased alternans vulnerability. A different theoretical
results of an earlier study (872) in mice. Ito is downregu- study also highlighted the importance of SR calcium
lated in chronic AF (7, 549), which is well reflected in the release dynamics and how their changes in heart failure
widening of early atrial repolarization of AF and its promote alternans (858).
increased AP duration at 20% repolarization (APD20) The ionic mechanisms underlying electrophysio-
Intracellular Ca21 handling is impaired in chronic AF logical remodeling in the ventricle include downregu-
associated with heart failure (893), as increased open lation of Kv4.3 channels and Ito, Kir2 channels and IK1,
probability of the ryanodine receptor SR channels SK2 channels, KvLQT1 channels, and IKs (156–159,
results in more frequent Ca21 release from the intracel- 866, 873). Peak INa can be reduced in failing heart by
lular Ca21 store (681, 824, 887). This may subsequently posttranscriptional reduction and deficient glycosyla-
enhance the frequency of spontaneous depolarization tion of Nav1.5 channel a-subunit (862), and these
in AF via NCX, possibly contributing to the increased changes may contribute to slowing of impulse and
triggered activity in chronic AF (803). conduction. In a mouse transverse aortic constriction
Structural remodeling has been observed in a tachy- HF model, Nav1.5 remodeling was observed in sub-
paced AF goat model (880), which seems even more cellular microdomains: Nav1.5 cluster size at the lat-
pronounced in AF associated with heart failure (894). eral membrane and at the intercalated disk was
This includes elevated levels of fibrosis (34) and other reduced in failing myocytes, in agreement with
extracellular matrix components such as myofibroblasts, reduced peak INa and impaired transversal and longi-
periostin, matrix metalloproteinases (MMPs), and trans- tudinal conduction in failing myocardium (98). More
forming growth factor b1 (TFG-b1). Kidney disease (895), importantly, late INa is significantly increased both in
inflammation (29), and oxidative stress (896, 897) were an experimental heart failure model (127) and in
also associated with arrhythmogenesis and remodeling patients (126); the substantial inward current during
in AF. Several reports proved the important role of cal- the plateau phase causes failure of repolarization
modulin-dependent protein kinase II (CaMKII) in both and decreases its homogeneity, providing substrate
abnormal Ca21 handling and structural remodeling in for reentry arrhythmias. In rabbit and human ventricular
AF, suggesting CaMKII inhibition as a promising new muscle, downregulation and change of the transmural
strategy to treat AF (573, 898, 899). expression pattern of CFTR Cl channels was observed
(906) in the hypertrophied heart. Connexin 43 protein
7.2. Remodeling in Heart Failure downregulation and its lateralization delay impulse con-
duction and can enhance anisotropic impulse propaga-
Sudden cardiac death caused by arrhythmias (900) is re- tion (878), further contributing to substrate generation
sponsible for a substantial proportion of the mortality in for arrhythmias.
heart failure. The overwhelming majority of published In addition to remodeling of ionic channels, cal-
papers confirmed significant lengthening of repolariza- cium handling is substantially altered, showing
tion of the ventricular muscle without substantial species increased SR leak and reduced SR reuptake, result-
differences and regardless of its origin. This is also asso- ing in low SR calcium loading and thus diminished
ciated with increased dispersion of repolarization (241), calcium transient (802). An additional factor contrib-
which can be further enhanced by bradycardia-induced uting to low SR loading and calcium transient is the
AP prolongation often seen in this situation. Enhanced increased NCX (907). The alterations of calcium

1128 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

handling affect the SR-mitochondria cross talk, lead- structural left ventricular remodeling (922). Indeed, dis-
ing to energy starvation of failing cells, affecting organized myocyte architecture (923), intramyocardial
energy-sensitive processes (908). CaMKII signaling and replacement fibrosis, and collagen deposition in
is also strongly affected, with relevance for arrhyth- regions with chronic microvascular ischemia (924) can
mia formation (802). serve as substrates for sustained arrhythmias (925). In
Changes in electrophysiology and calcium handling in addition, the surrounding ischemic myocardium mani-
heart failure promote ectopic automaticity in the PVs, fests impaired conduction and heterogenous repolariza-
atria, and ventricle, serving as an important trigger for tion dispersion in advanced stages of HCM (926).
reentry arrhythmias (788, 790, 875, 909, 910). One type Alternans is another source of proarrhythmic substrate
of automaticity may arise from pacemaker-like currents in HCM (927). However, most SCD events occur in HCM
in ventricles. The pacemaker HCN4 and HCN2 channels patients at early disease progression stages, when left
that carry the If inward pacemaker current have been ventricular hypertrophy is the only major structural ab-
reported to be upregulated in ventricular (788, 789, 875, normality (928). Accordingly, several key elements and
876, 911) and atrial (912) tissue of heart failure patients. In consequences of electrical remodeling have been iden-
contrast, downregulation of If in the SAN has also been tified in cardiomyocytes from HCM patients, including
observed (913). A second type of automaticity may arise more frequent EAD and DAD formation (160), increased
from afterdepolarizations resulting from increased cal- diastolic Ca21 concentration and elevated NCX expres-
cium leak and subsequent spontaneous SR release, to- sion (160), and increased left ventricular HCN4 expres-
gether with the upregulated NCX (907). This increased sion (792), all capable of providing arrhythmia triggers in
risk of DADs is further enhanced by the above-men- HCM (27, 929, 930). In addition, prolongation of the
tioned reduction in IK1 (291), which makes it easier for the APD (160) and, consequently, QTc lengthening were
depolarization to reach the threshold of INa reactivation. observed in patients with HCM, which can be caused by
More importantly, heart failure alters cellular Ca21 han- the reduced densities of Ito and IK1, decreased expres-
dling (681, 914). Calcium leak from the SR is enhanced, sions of hERG1b and KCNQ1, and increased INaLate and
resulting in spontaneous Ca21 release from the SR slower ICa,L inactivation kinetics (160, 931) in myocytes
(802, 824, 915). This transient intracellular Ca21 concen- from HCM patients. These elements of ionic current
tration increase activates the already upregulated (907) remodeling were sufficient to recapitulate increased vul-
forward NCX, leading to arrhythmogenic depolarizations nerability to EADs when incorporated into a ventricular
serving as triggers in an environment where the sub- myocyte model (776).
strate also favors the development of arrhythmias due In competitive athletes, chronic endurance training
to heterogeneity of impulse conduction and repolariza- leads to the development of the so-called athlete’s
tion. In this process, CaMKII seems to have an important heart, characterized by lower resting heart rate due to
role since its inhibition can have beneficial antiarrhyth- increased vagal tone (932), If downregulation (933), and
mic consequences (802). symmetric cardiac hypertrophy (934). The incidence of
sudden cardiac death in top competitive athletes is two
7.3. Remodeling in Hypertrophic Cardiomyopathy to four times higher compared with the age-matched
and in Athlete’s Heart population (935). Interestingly, hypertrophic cardiomy-
opathy is found in almost 40% of the cases of SCD in
Hypertrophic cardiomyopathy (HCM) was first described athletes (919). The electrophysiological mechanisms
in 1958 after autopsies on individuals who suffered underlying SCD in athletes are not known (for a review
unexpected sudden cardiac death (916). Since then, see Ref. 18); nevertheless, a significantly larger short-
HCM has been identified as the most common inherited term variability of the QT interval was found in professio-
heart disease, with a prevalence of 1:500 (917), caused nal soccer players, indicating increased repolarization
by >1,400 known mutations in at least 11 genes (most temporal instability (936) that can refer to increased ar-
commonly in b-myosin heavy chain, MYH7 and myosin rhythmia susceptibility in this population (259, 937).
binding protein C, MYBPC3 genes) encoding different
sarcomeric and Z-disk proteins (243). The disease is 7.4. Remodeling in Diabetes Mellitus
characterized by variable left ventricular hypertrophy
and fibrosis (918) and increased incidence of serious Diabetes mellitus represents a significant burden on
ventricular arrhythmias (919, 920). Moreover, HCM is the health care systems all over the world since its preva-
most common cause of SCD in young persons and com- lence is continuously increasing. Diabetes mellitus, an
petitive athletes under the age of 35 yr (919, 921). The endocrine disorder, is characterized by reduced insulin
increased propensity for ventricular arrhythmia develop- production (type 1) or increased insulin resistance (type
ment in patients with HCM is mainly attributed to 2), with the latter belonging to cardiometabolic disorders

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1129


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

and being responsible for 90% of diabetes mellitus enhanced CaMKII-mediated phosphorylation of RyR2
cases. Cardiac repolarization disturbances like QTc has been linked to aberrant Ca21 handling in the same
lengthening or increased QT dispersion are associated animal model (949, 950). Although it is less well estab-
with the disease, and cardiovascular complications lished, similar pathophysiological alterations have been
including arrhythmias and sudden death are major observed in type 2 diabetic rats (951, 952). Results
causes of mortality and morbidity in diabetes. These obtained from experimental diabetes suggest that the
complications usually develop slowly over several deca- diabetic heart provides favorable substrate and inc-
des and are associated with cardiomyopathy, ischemia, reased propensity of trigger for arrhythmias due to
vascular sclerosis, and neuropathy with complex and enhanced ectopic activity and both impaired impulse
not fully understood mechanisms. Here, we briefly conduction and repolarization, but results are not con-
summarize those electrophysiological and structural clusive. Therefore, further studies with more appropriate
changes that can be attributed directly to diabetes- experimental diabetes models are needed to better
induced remodeling (938). The majority of the available understand the mechanisms of arrhythmias in diabetes
data regarding electrical remodeling are derived from mellitus.
type 1 diabetes animal models. There, diabetes is
induced acutely by alloxan or streptozotocin (raising the 7.5. Myocardial Ischemia, Infarction, and
question of how well the model corresponds to chronic Arrhythmias
changes developing slowly over decades in humans).
Results in type 1 diabetes animal models include length- Myocardial ischemia mostly develops on the basis of
ening of ventricular repolarization, attributed to down- coronary artery disease following critical decreases in
regulation of Ito (868) and IKs currents. At the level of blood flow in obstructed coronary arteries. Occlusion of
mRNA and protein, Kv4.2, Kv4.3, and minK were reduced coronary arteries induces a chain of events within
but Kv1.4 and KvLQT1 did not change or were upregu- minutes of the onset of ischemia, leading to altered func-
lated. Zhang et al. (869, 939) reported significant length- tion of ion channels and concomitant ischemia- and in-
ening of QTc and downregulation of cardiac HERG farction-induced cardiac arrhythmias. The majority of
channels and IKr in alloxan-induced type 1 rabbit diabe- cases where ventricular tachyarrhythmias including ven-
tes; however, the latter results regarding IKr were not tricular fibrillation lead to sudden cardiac death are
confirmed by other studies in rabbits and dogs (271, clearly associated with coronary artery disease and
867). In Goto–Kakizaki type 2 diabetic rats, ICa,L and myocardial infarction (953, 954). Reperfusion of the
Ca21 transient did not change compared with those in myocardium is necessary for tissue survival; however, it
the control animals but decreased atrial KCNN2 mRNA, can also lead to reperfusion-induced arrhythmias (955,
KCa2.2 protein levels, and corresponding SK2 current 956). When timely revascularization is not performed,
densities were observed (874, 940), with concomitantly coronary obstruction leads to irreversible cell damage
enhanced myocardial hypertrophy and extracellular ma- termed myocardial infarction, and the dead cardiomyo-
trix deposition with fibrosis (941). In Zucker rats, another cytes are replaced by scar tissue. The remodeling in sur-
type 2 diabetic model, enhanced fibrosis, Cx43 laterali- viving epicardial and border zone myocytes mostly
zation, and significant APD prolongation with delayed leads to conduction abnormalities, while surviving endo-
ICa,L inactivation (942) were observed (865). Another ob- cardial Purkinje fibers can be important sources of trig-
servation in type 1 and 2 rat diabetes models is the gered activity (618). In addition, over the course of days
reduced impulse conduction reserve (863, 943). In a and weeks, the noninfarcted myocardium, including the
recent paper, O-GIcNAcylation and impaired function of peri-infarct border zone, also undergoes significant
Nav1.5 channel (864) and INa were described, which to- remodeling that favors arrhythmia development due to
gether with the fibrotic structural changes (944) can also changes in conduction, refractoriness, and triggered ac-
contribute to the arrhythmogenic impulse conduction tivity (31). An additional source of increased dispersion
defects (945). of refractoriness is the vulnerability of the infarct border
In addition to changes in ionic channels, calcium han- zone to alternans, shown both computationally (847)
dling is also altered. Several experimental studies in and experimentally (847). Such spatially localized alter-
both type 1 and 2 diabetes models in rat established nans is proarrhythmic in a fashion similar to spatially
increased SR Ca21 leak due to enhanced RyR2 channel discordant alternans, forming steep gradients of repola-
activity (946, 947), which, as mentioned above, would rization. Multiple other studies have investigated arrhyth-
be expected to result in DADs and extrasystoles. mia mechanisms associated with myocardial ischemia
Accordingly, Yaras et al. (948) showed increases in and infarction; for comprehensive detailed reviews, the
Ca21 spark frequency in cardiomyocytes from RYR2 reader is referred to works by Janse and Wit (31) and
and decrease in FKBP12.6 (calstabin) association. Also, Pinto and Boyden (784).

1130 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

In the acute phase of myocardial ischemia (within potentials between neighboring ischemic (depolari-
minutes to 2–4 h after coronary blood flow reduction), zed) and normoxic (hyperpolarized) myocardium and
ventricular arrhythmias including fibrillation occur in has been shown to contribute to ectopic activity in ex-
humans (957) and also in experimental animals sub- perimental conditions (972–974) and computer simu-
jected to complete coronary artery occlusion (958, 959). lations (706). In general, the initiation of so-called
Acute ischemia promotes the formation of arrhythmia “phase 1b” arrhythmias in experimental models of
substrate, which allows reentry via several mechanisms. myocardial ischemia is mostly attributed to increased
The resting membrane potential of ischemic cardiomyo- automaticity (31).
cytes is significantly depolarized (960), partly due to In the subacute and chronic phases of myocardial in-
marked K1 loss and intracellular acidosis (783), leading farction, reentry and triggered activity caused by early
to extracellular K1 accumulation in ischemic tissue (961). and delayed afterdepolarizations are major mecha-
The depolarization of the resting membrane potential nisms of infarction-induced ventricular arrhythmias
depresses action potential upstroke and amplitude by (28, 975), where surviving Purkinje fibers (976) and epi-
reducing INa and results in the prolongation of the refrac- cardial border zone myocytes (977) play a critical role
tory period via slow recovery from inactivation of the so- in arrhythmia development. A series of papers by the
dium gates (39). Activation of ATP-dependent potassium Boyden group describe the morphological and electro-
channels shortens the action potential duration (962). physiological changes occurring over 24–48 h after an
Within 15 min of the onset of ischemia, gap junction infarction in surviving left ventricular Purkinje cells that
channel phosphorylation status is changed and they get underlie subacute spontaneous arrhythmias (for a
translocated into intracellular pools in ischemic cells detailed review, see Ref. 784). In general, surviving
(963), severely reducing cell-to-cell coupling (815, 964, Purkinje fibers exhibit depolarized resting potentials
965). These changes result in impaired impulse conduc- associated with reduced IK1 (978), decreased ICa,L and
tion in acute ischemia (966). Increased spatial dispersion ICa,T densities (979), prolonged repolarization with up
of repolarization and refractoriness between different to 51% smaller Ito density, and delayed Ito reactivation
regions of the myocardium is also an important arrhyth- kinetics (980). Also obs-erved were nonuniform cal-
mia substrate (128). Nonuniform shortening of the action cium transients and abnormal calcium waves leading
potential due to heterogeneous sarcolemmal IK,ATP acti- to spontaneous action potentials and triggered activity
vation in myocytes from the ischemic and border zone in the infarcted heart (619). The surviving epicardial
(reviewed in Ref. 967) and no shortening of AP in the myocytes of the border zone next to the infarcted area
normoxic myocardium increases dispersion of repolari- exhibit decreased excitability and resting potential,
zation and effective refractory period within minutes af- reduced upstroke velocity and amplitude of the action
ter ischemia onset (340). The first phase of ventricular potential (977), and, importantly, marked postrepolari-
arrhythmias (“phase 1a”) peaking between 5 and 6 min zation refractoriness (32, 981) due to decreased INa
after the onset of ischemia in experimental models is density and altered INa kinetics (982). A pathological
usually, but not exclusively, attributed to the mecha- redistribution of connexin 43 gap junction protein and
nisms described above (31, 968), which have been fur- reduced gap junctional conductance was observed in
ther investigated in computer simulation studies using border zone surviving cardiomyocytes (983). These
human biventricular models (969). changes lead to abnormal, slow, and anisotropic
Multiple mechanisms underlie the formation of ar- impulse conduction (28, 984, 985). Interestingly, in
rhythmia triggers in acute ischemia. The increased these surviving epicardial myocytes, a gradual short-
release of catecholamines occurs 15–20 min after the ening and triangularization of the action potential was
onset of myocardial ischemia (970), possibly increasing observed over 2 wk, followed by a return of action
ectopic activity. Early and delayed afterdepolarizations potential parameters to normal by 2 mo after myocar-
can supply the triggers for arrhythmia initiation in acute dial infarction (986). The reduced ICa,L density (987)
myocardial ischemia. Early afterdepolarizations can de- may be partly responsible for the triangularization of
velop in Purkinje fibers in acute ischemia because of in- the action potentials on these cells. Several important
tracellular acidosis and exposure to ischemia-induced repolarizing K1 currents are downregulated in surviv-
release of lysophosphatidylcholine (971). Delayed after- ing epicardial cells, including Ito (977) and IKr and IKs
depolarizations can occur in ischemic cells because of (240), leading to prolonged action potentials. These
acidosis, hypoxia, and calcium overload (617). A so- changes described above in epicardial border zone
called “injury current” flowing from ischemic myocar- myocytes strongly favor the development of reentry
dium to normal myocardium can cause increased abnor- and, of particular interest, anisotropic reentry (7).
mal automaticity in acute myocardial ischemia. The In addition to the changes described above, the
basis for this current is the difference in membrane remote noninfarcted regions of the myocardium also

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1131


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

exhibit structural and electrical remodeling as the responsible for repolarization prolongation in aged
remaining myocardium in the infarcted heart adapts to hearts (1009, 1010). These elements of ventricular
increased workload (988) The developing regional left remodeling in the aged heart increase susceptibility to-
ventricular hypertrophy is an important risk factor for the ward arrhythmias by altering cellular coupling, increasing
generation of severe ventricular arrhythmias (989) and anisotropic conduction, and enhancing heterogeneity of
probably relates to enhanced dispersion of repolariza- refractoriness in the myocardium, which are changes
tion associated with myocardial hypertrophy. that all together favor reentry initiation and stabilization.
The increased expression of NCX, the delayed inactiva-
tion of ICa,L, together with reduced expression of SERCA
7.6. Aging-Associated Cardiac Remodeling and other proteins related to calcium handling all con-
tribute to impaired calcium homeostasis and provide the
There is a dramatic increase in the incidence and preva- mechanisms for increased triggered activity in the aging
lence of cardiovascular disease and mortality with age, myocardium (1011–1015).
and aging is now identified as a major risk factor for cardi- The ability of the heart to properly respond to auto-
ovascular morbidity (990). Evidence suggests that cardio- nomic stimuli also decreases with aging (1016). Red-
vascular aging is associated with remodeling of the heart, uced b-adrenoceptor responses and receptor den-
setting the stage for arrhythmia development (especially sities were identified in the aging myocardium (1017,
atrial fibrillation and ventricular tachycardia) and reduced 1018), exacerbated by decreased cAMP production
cardiac function. The incidence of sudden cardiac death (995). It is not clear how impaired cAMP-dependent
increases with age (991); however, our understanding of regulation of IKs (1019) and ICa,L (1020) translates into
the mechanisms responsible for increased incidence of alterations of repolarization reserve in the elderly. In
arrhythmias in the elderly remains limited (992). Many of addition, decreased vagal component of heart rate
the aging-associated alterations are, at least in part, inevi- variability and heart rate responses were observed af-
tably due to the observed coronary artery dysfunction, ter muscarinic acetylcholine receptor blockade (1021,
increased artery stiffness, decreased responsiveness to 1022). The impaired autonomic regulation of the heart
b-adrenergic stimulation, and cardiac extracellular matrix and the electrical and structural remodeling described
remodeling found in the elderly (993–995). above in the elderly contribute to reduced cardiac
The pacemaker function of the sinus node signifi- adaptive responses and increased supraventricular
cantly decreases in older patients, probably because of and ventricular arrhythmia susceptibility (1023).
replacement of pacemaker cells within the sinoatrial
node (SAN) by extracellular matrix (996). Animal studies
suggest that decreased expression of HCN2, ICa,L, ICa,T, 8. INHERITED CONDITIONS ASSOCIATED
and Kv1.5 channels can also contribute (548, 997, 998). WITH ION CHANNEL DYSFUNCTION
Consequently, the rate of spontaneous diastolic depola-
rization falls and action potential duration is prolonged Cardiac arrhythmias are usually symptoms or consequen-
in pacemaker cells (523, 999). The intrinsic heart rate ces of other underlying diseases such as myocardial is-
decreases (997), with concomitant fatigue, bradycardia, chemia or infarction, heart failure, hypertension, diabetes,
and increased incidence of supraventricular arrhythmias etc. However, cardiac ion channel mutations or polymor-
including atrial fibrillation in the elderly (1000). phisms (1024) can represent the primary cause of arrhyth-
The observed delayed and impaired cardiac impulse mias, collectively called “ion channelopathies” (1025).
conduction, with concomitant QRS complex widening, is This field is particularly rapidly expanding, and detailed
the result of decreased Cx43 expression and degenera- discussion is far beyond the scope of this review; some
tive changes in the cardiac connective tissue (992, excellent recent reviews are suggested for further read-
1001–1003). ing (114–117, 149, 246, 1026–1029).
In aged ventricles, even without the presence of struc- In general, mutation of a certain ion channel can
tural heart disease, loss of cardiomyocytes, reactive hy- cause gain or loss of function (1029–1032) resulting in
pertrophy in the left ventricle, fibrosis, and changes in increased or decreased current through the affected ion
repolarization occur (161, 1004–1006). The prolongation channel, which may alter arrhythmia trigger and/or sub-
of the ventricular action potential was observed in sen- strate (TABLE 1) (245, 1033, 1034).
escent sheep (1007), and consequent QT interval length- Congenital LQT syndrome is characterized by prolon-
ening was shown in old dogs (1007, 1008). In an attempt gation of myocardial AP and QT interval caused by the
to maintain cardiac function, ion channel remodeling reduction of repolarizing current due to either loss-of-
involving ICa,L, Ito, KATP expression changes, delayed ICa,L function mutations in repolarizing current-conducting
inactivation, and increased INa,late are probably potassium channels or gain-of-function mutations in

1132 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

depolarizing current-conducting sodium or calcium channels, i.e., KCNH2 (SQT1), KCNQ1 (SQT2), and KCNJ2
channels (114, 115) (TABLE 1). Congenital LQT syndrome (SQT3), have been associated with the disorder (1046–
is commonly associated with the development of TdP 1048) (TABLE 1). In addition, loss-of-function mutations in
and sudden cardiac death (1035), and most LQT syn- genes encoding voltage-gated calcium channel subunits
drome subtypes show an autosomal dominant inheri- CACNA1C, CACNB2b, and CACNA2D1 were designated
tance (115). In >90% of patients with congenital LQT, as SQT4, SQT5, and SQT6, respectively (1049) (TABLE 1).
genetic variants of KCNQ1 encoding the a-subunit Kv7.1 Patients with SQT syndrome involving calcium channel
of IKs (LQT1; 30–35% of cases), KCNH2 encoding the mutations exhibit an overlapping phenotype combining
a-subunit Kv11.1 of IKr (LQT2; 25–40% of cases), and short QT and a Brugada syndrome phenotype (1050).
SCN5A encoding the a-subunit Nav1.5 for INa (LQT3; 5– Loss-of-function mutations in the gene (SLC22A5) encod-
10% of cases) underlie the disorder (122, 1036, 1037) ing a sodium-dependent carnitine transporter have been
(TABLE 1). A number of other genes encoding ion chan- termed SQT7 (1051) (TABLE 1). In SQT7, the mechanistic
nel proteins or their regulators have been implicated as link between short QT and carnitine concentrations
causes of congenital LQT syndrome (TABLE 1). For remains elusive; however, increased IKr due to the lack of
details on the causative roles of the genetic variants long-chain acylcarnitines may play a role (1052). SQT8 syn-
involved in the different congenital LQT syndrome sub- drome involves a mutation in the gene (SLC4A3) encoding
types, the reader is referred to other comprehensive a Cl /HCO3 exchanger (AE3) leading to a trafficking defect
reviews (115, 246). In LQT syndrome, prolongation of of the mutated AE3 to the cell membrane (1053) (TABLE 1).
repolarization and refractoriness, accompanied with The shortening of APD and ERP in the myocardium, as well
increased dispersion of repolarization (1038, 1039), as the increased spatial dispersion of repolarization, due to
enhance the substrate for typical ventricular arrhythmias more enhanced APD shortening in the epicardium, all favor
in patients with LQT syndrome such as TdP polymorphic reentry tachycardia development both in the ventricles and
tachycardia and ventricular fibrillation. Enhanced trig- in the atria in SQT syndrome (1054, 1055).
gered activity due to early afterdepolarizations, elicited Brugada syndrome (BrS) (1056) and arrhythmogenic
by reactivation of ICa,L during the prolonged AP, provides right ventricular cardiomyopathy (ARVC) (1057) are
the trigger for the development of TdP in LQT syndrome inherited cardiac diseases with a complex genetic back-
(796, 1038). LQT syndrome is characterized by incom- ground. Brugada syndrome is characterized by ST seg-
plete penetrance and can be “silent,” i.e., no significant ment elevation in right precordial leads and has been
prolongation of repolarization and QT interval is considered as the cause of SCD in up to 20% of patients
observed. However, congenital LQT syndrome greatly with a structurally normal heart (778). Approximately 20–
enhances the effect of any other factor which delays 30% of patients with BrS exhibit pathogenic mutations in
repolarization by other mechanisms. This can be the SCN5A gene encoding the a pore-forming subunit
explained by the concept of “repolarization reserve,” as of the cardiac Nav1.5 sodium channel (1058), leading to
introduced in sect. 3.4.2 (228, 257, 1040, 1041). loss of function in fast INa and, consequently, conduction
Accordingly, impaired function of different types of po- slowing and delayed right ventricular activation (1059,
tassium channels (caused by genetic mutation, disease- 1060). In BrS, because of decreased INa the balance of
induced downregulation, or drug effects) can add to- outward and inward currents changes and the repolariz-
gether, further enhancing the substrate for ventricular ing effect of Ito is amplified in epicardial but not in endo-
arrhythmias. Of note, ion channel polymorphisms can cardial cells, resulting in enhanced transmural dis-
also play a role in arrhythmia development, thus empha- persion of repolarization. This increased dispersion can
sizing the importance of pharmacogenetics during the create a vulnerable window for extrasystoles to induce
evaluation of proarrhythmic drug-induced adverse phase 2 reentry arrhythmias (1061, 1062). In a genome-
effects (798, 1024). Which current is affected by such wide association study, genetic variants of SCN10A-
polymorphisms (usually potassium currents, ICaL, or encoded Nav1.8 sodium channels, playing a key role in
INaLate), may largely determine the individual’s response cardiac neurons (1063) but also expressed in the work-
to antiarrhythmic channel-blocking drugs, emphasizing ing myocardium (1064), have been identified as strong
the concept of personalized med-icine. modulators for BrS (1065). In addition, in 2–5% of BrS
SQT syndrome (1042, 1043) is a rare, albeit severe, chan- patients pathogenic variants have been identified in a
nelopathy that is characterized by abnormally short fre- number of genes encoding other ion channel proteins,
quency-corrected QT intervals (<360 ms), due to and up to 70% of patients with BrS have an unidentified
accelerated ventricular repolarization (1044), with a high genetic cause (778, 1058). Interestingly, the expression
risk of sudden cardiac death and AF (1045). SQT syndrome of Kv4.3, a key subunit of Ito (135), was markedly reduced
shows autosomal dominant inheritance, and gain-of-func- in the endocardium but not in the epicardium in patients
tion mutations in genes encoding different potassium with BrS (1066). This enhanced transmural difference in

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1133


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

Ito can lead to increased transmural dispersion of repola- 9. OTHER FACTORS INFLUENCING
rization in BrS patients, favoring the development of ARRHYTHMOGENESIS
reentry arrhythmias (24, 1062).
In patients with ARVC, a significant number of cardio- 9.1. Sex Differences in Cardiac Electrophysiology
myocytes in patches of the right ventricular free wall are and Arrhythmias
replaced by fibroadipose tissue, with marked patho-
physiological consequences for myocardial depolariza- Sex differences in cardiac electrophysiology carry
tion and repolarization of the right ventricle (1057). The marked clinical significance because they translate into
causative role of mutations in at least 16 genes, mostly distinct arrhythmia risks and outcome in men and women.
encoding different desmosomal proteins, has been Although women have a somewhat smaller risk of AF or
identified in ARVC (1067), whereas in 40–50% of ventricular fibrillation (VF) compared with men (1081), this
patients the genetic cause is unknown (1068). The exact does not hold for all causes of arrhythmia. For example,
molecular mechanisms for how desmosome assembly women have a longer frequency-corrected QT interval
impairment and dysfunction lead to cardiomyocyte apo- and are more susceptible to development of drug-
ptosis and their fibroadipose replacement are not fully induced polymorphic ventricular tachycardia (1082–
understood (1069, 1070). ARVC is associated with fre- 1085), whereas men are more likely to exhibit early repo-
quent ventricular extrasystoles, ventricular tachycardia larization (150). Here we briefly discuss the underlying
with left bundle branch block morphology, syncope, and mechanisms for sex differences in cardiac electrophysiol-
SCD (1071). On autopsy, ARVC is found to be responsible ogy; for more detailed discussions the reader is directed
for SCD in 5% of young competitive athletes in the to recent comprehensive reviews (265, 1081).
United States, whereas this number is around 27% in Sex hormones exert their effects via both transcrip-
northern Italy (1072, 1073). The fibroadipose tissue tional regulation (1086, 1087) and acute, nongenomic
patches correspond to electroanatomical scars that pro- modulation of intracellular signaling (1088). Women ex-
mote the establishment of scar-related macro reentry hibit greater sinoatrial node automaticity (1089), and
circuits (1074, 1075). pregnancy increases pacemaker If current density along
Catecholaminergic polymorphic ventricular tachycar- with HCN2 expression in mice (1090). A larger INaLate
dia (CPVT) is characterized by a normal resting ECG; was observed in male rabbit left atrial posterior wall
however, malignant ventricular arrhythmias and SCD are myocytes (1091). Since these cells are major sources of
induced by adrenergic stimulation (821, 1076). Mutations non-pulmonary vein triggered activity (204, 1092), and
in RYR2, encoding the ryanodine receptor 2, the Ca21 INaLate is an important contributor in atrial arrhythmogen-
release channel located on the SR (autosomal dominant esis (1093), the larger INaLate may contribute to higher
CPVT type 1), and in CASQ2, encoding calsequestrin 2, triggered activity in males, leading to the initiation of AF.
the calcium binding protein inside the SR (autosomal re- The prolonged ventricular repolarization in women is
cessive CPVT type 2), makes these channels and cal- partly due to decreased expression of pore-forming
cium storage sites more sensitive to catecholamine- and/or auxiliary subunits of ion channels carrying repola-
induced Ca21 release (1077). Consequently, spontane- rizing currents, including Ito, IK1, IKr, IKs, and KATP in
ous triggered automaticity is induced by depolarizations humans (151), and these results are generally in agree-
due to increased transient inward current elicited by for- ment with animal studies showing lower densities for
ward NCX activity during diastole. Iyer et al. (1078) used these currents (263, 1094–1096). In addition, 17b-estra-
computer simulations to characterize the impact of diol (E2) directly (nongenomic effect) reduces IKr and
RYR2 and calsequestrin mutations in the CPVT pheno- enhances IKr block and QTc prolongation by HERG
type. Their study shows how impaired SR calcium sens- blocker drugs (1085, 1097), whereas testosterone
ing and increased spontaneous SR calcium release increases IKr and IKs (1094, 1098) and progesterone
promote DADs and how these events are aggravated enhances IKs (1099). Indeed, the normalization of the
by b-adrenergic stimulation. QTc interval in a woman with long QT syndrome was
Mutations in If pacemaker channels can cause both observed during pregnancy due to increased progester-
brady- and tachycardias (1034) by altering sinus automa- one levels (1100). A greater transmural IKs density gradi-
ticity. Sinus bradycardia was also reported in patients ent was found in female dogs compared with males
with loss-of-function mutation of the SCN5A sodium (1101), possibly contributing to larger transmural APD het-
channels resulting in failure of pacemaker capture erogeneity in females. Opposite effects of b-adrenergic
(1079). Some familial AF cases are also linked to different stimulation by isoproterenol on Purkinje fiber APD were
ion channel mutations in the atria, influencing repolariza- observed in male dogs (APD shortening) compared with
tion, depolarization, connexin function, and conse- female dogs (APD prolongation), highlighting sex- and
quently impulse conduction properties (1080). age-related differences in the autonomic regulation of

1134 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

the cAMP-dependent IKs current (1102, 1103). Since IKs is above, extracellular K1 concentration significantly mod-
a key current for repolarization reserve (228, 250, 260), ulates several K1 currents (225, 233–235, 294, 1114) in
these changes may lead to reduced repolarization the range of blood K1 concentrations (2–10 mM)
reserve and an increased arrhythmia substrate in observed frequently in clinical practice. Hyperkalemia
women. depolarizes the cell, as expected from the Nernst equa-
A more heterogeneous transmural INa distribution was tion. The depolarization caused by elevated extracellular
found in female canine left ventricle (1104), and testoster- [K1] can also slow impulse conduction indirectly, by
one increased epicardial INa amplitude in female dogs to decreasing IK1 and partially inactivating inward currents.
levels similar to those measured in male epicardium This happens during ischemia and when K1 accumula-
(1104). As INa and Ito have opposing effects on repolariza- tion occurs in the intercellular clefts, not immediately
tion, increased transmural heterogeneity in ion channel handled by the Na1-K1 pump. This latter mechanism
expression creates transmural dispersion of repolariza- may contribute to frequency-dependent regulation of
tion that serves as an arrhythmogenic substrate (128). APD and conduction as well (4). IKr is increased by eleva-
Sex-dependent differences in ICa,L densities and Ca21 tion of extracellular K1 concentration and decreased in
homeostasis protein expression and regulation (162) hypokalemia (235, 294, 1115), which is not what would be
lead to sex differences in triggered activity, since the expected from its transmembrane concentration gradient
reactivation of the L-type calcium current during the pla- and is explained by hypokalemia-induced acceleration of
teau of the action potential and Ca21 cycling protein rapid IKr inactivation (225). This may have a role in fre-
dysfunction importantly contribute to EAD formation (10). quency-dependent APD regulation, and it helps in unde-
Larger ICa,L densities were found in all transmural layers rstanding why hypokalemia enhances the risk of arrhyth-
of the left ventricle in female dogs (1101), possibly con- mias by causing prolongation of repolarization at slow
tributing to a bigger transmural heterogeneity of APD in heart rate. IKs, unlike IKr, is reduced by elevated [K1]o and
females, since IKs (similar in male and female midmyocar- increased by hypokalemia (235, 294). Therefore, IKs may
dium) could not offset the increased ICa,L. In addition, es- serve as part of the repolarization reserve in hypokalemic
tradiol increased ICa,L (1105) and promoted EAD patients. Hypokalemia and diminished IK1 can decrease
formation and sudden cardiac death in rabbits with background K1 currents, which may favor ectopic auto-
LQT2 syndrome (1106), whereas testosterone and pro- maticity not counterbalancing existing inward currents
gesterone exerted opposite effects (1088, 1106, 1107). In such as If or NCX and may act as enhanced triggers for
female rabbit left ventricle, NCX expression and current arrhythmia development. Hypokalemia was shown to
were higher, resulting in increased EAD development decrease Na1-K1 pump activity, thereby elevating intra-
following the administration of the HERG blocker dofeti- cellular [Na1] and leading to increased DAD formation
lide (1108). Testosterone, however, increased SERCA via enhanced NCX function (1116). Hypokalemia also
function, causing rapid removal of excess intracellular decreases IKr and consequently prolongs repolarization
Ca21 (1109). Testosterone increased RyR2 activity (1109), (225, 235, 1117), decreases repolarization reserve, and of-
whereas estradiol increased RyR2 leakiness and con- ten leads to torsade de pointes tachycardia, especially
tributed to the formation of proarrhythmic afterdepolari- when K1 channels are already inhibited by concomitant
zations (1110). A significantly lower expression of Cx43 drug therapy. Hypomagnesemia has a similar effect but
was found in left ventricular myocardium in women com- with a different mechanism. The Mg21 ion acts as endo-
pared with men (151) that can make women more sus- gen Ca21 antagonist, and in the case where its serum
ceptible to ventricular conduction impairment. level is low it cannot fine-tune the influence of ICa,L in
In summary, sex hormones significantly modulate sinus node function, repolarization, and possible EAD
both the arrhythmia substrate and triggered activity and DAD formation (1118).
leading to sex differences in ECG morphology and sus-
ceptibility to supraventricular and ventricular arrhyth- 9.3. Proarrhythmic Drug Effects
mias. Estradiol lengthens repolarization, promotes EAD
formation, and exacerbates the QT-prolonging effect of Not only can diseases, remodeling, serum electrol-
HERG blocker drugs, leading to higher incidence of car- yte disturbances, and genetic disorders contribute to
diac arrhythmias in females, whereas testosterone and arrhythmogenesis, but so can drugs, i.e., they may pos-
progesterone exert mostly opposite effects. sess proarrhythmic actions. Positive inotropic drugs like
digitalis, which inhibits the Na1-K1 pump and enhances
9.2. Serum Ion Concentration Changes Ca21 release of SR, can cause DADs, whereas phospho-
diesterase inhibitors like milrinone or sympathetic s
Potassium is by far the most important ion in blood that timulators (1119) like amphetamine, epinephrine, and nor-
affects arrhythmogenesis (233, 1111–1113). As mentioned epinephrine increase intracellular cAMP levels (1120,

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1135


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

1121), and they consequently increase If-related and management and prevention of underlying diseases are
Ca21 overload-induced automaticity; all of these repre- needed. This requires a better understanding of the car-
sent enhanced triggers for arrhythmias. A large number diac electrophysiological function and how it may be
of drugs inhibit INa and impulse conduction including modulated. The latter includes the improved understand-
class I antiarrhythmic drugs like quinidine, flecainide, ing of the nature of cardiac action potentials in various
propafenone, and tricyclic antidepressants (202, 1122, regions of the heart and the function of the underlying
1123). These drugs, despite the fact that some of them transmembrane ionic currents, pumps, and the develop-
are used to abolish arrhythmias, can also elicit ventricu- ment of novel techniques and tools like in silico modeling.
lar tachyarrhythmia (1124, 1125), presumably by convert- Pathophysiological changes in the expression and func-
ing areas in damaged tissue into areas with tion of transmembrane currents and pumps lead to distur-
unidirectional impulse conduction block, which is one bances of impulse formation, conduction, and refract-
prerequisite of reentry arrhythmias, i.e., enhancing the oriness of cardiac muscle. These would favor the onset of
substrate for arrhythmogenesis. “triggers” and providing the “substrate” for these arrhyth-
Another proarrhythmic drug effect is prolongation of mias to develop. Therefore, further intensive research is
ventricular repolarization (1126). This proarrhythmic mech- required to improve our understanding of the physiology,
anism, which was discussed above, can enhance both pathophysiology, and genetic and hormonal modulation
the arrhythmic triggers by inducing EADs and the sub- of these ion channels and pumps in order to develop
strate by enhancing dispersion of repolarization. A con- more efficacious treatment modalities, thereby saving mil-
stantly growing number of drugs have such effects, such lions of lives in the future.
as class III antiarrhythmics like sotalol and ibutilide, antibi-
otics like erythromycin, antihistamines like terfenadine
and astemizole, antidepressants like sertindole, and anti- CORRESPONDENCE
malarial drugs like dihydroartemisinin and piperaquine
(1127). Most of these drugs had been identified to inhibit  (varro.andras@med.u-szeged.hu).
A. Varro
IKr or HERG current; therefore, the assessment of HERG
current-blocking properties of drug candidates became
mandatory in cardiac safety testing in drug development ACKNOWLEDGMENTS
(1128). It must be emphasized that some drugs can induce
We sincerely thank Gabriella Baczko and Dr. Noe
 mi To
 th for
repolarization abnormality-related arrhythmias without
apparent repolarization prolongation by impairing repola- administrative work on the manuscript.
rization reserve (260). Therefore, drug effects on other
repolarizing currents such as IKs, IK1, and Ito should also be
considered to avoid possible proarrhythmic complica- GRANTS
tions of novel compounds in development. Computer
This work was supported by the National Research,
simulation studies have recently demonstrated the impor-
Development and Innovation Office [NKFIH-K-119992, and
tance of evaluating multichannel effects for the prediction
GINOP-2.3.2-15-2016-00006 (MOLMEDEX) to A.V., NKFIH-K-
of drug-induced arrhythmic risk (715).
128851 to I.B., PD-125402, FK-129117 to N.N.], the János Bolyai
For a more detailed overview of proarrhythmic
Research Scholarship of the Hungarian Academy of Sciences
adverse effects of drugs, we refer the reader to Refs. to N.N., and the Hungarian Academy of Sciences, as well as a
108, 597, 1129–1132. Wellcome Trust Fellowship in Basic Biomedical Sciences
(214290/Z/18/Z) and an NC3Rs Infrastructure for Impact Award
(NC/P001076/1) to B.R.
10. CONCLUSIONS

Cardiac arrhythmias such as VF, TdP, and AF still DISCLOSURES


threaten the lives of millions of patients worldwide.
Although the underlying causes of arrhythmia develop- No conflicts of interest, financial or otherwise, are declared by
ment are variable, including ischemia, heart failure, the authors.
HCM, diabetes, extreme endurance training, or genetic
causes, the direct mechanisms leading to these arrhyth-
mias depend on changes of cellular electrophysiological AUTHOR CONTRIBUTIONS
properties or function of the heart. Therefore, to prevent
or treat cardiac arrhythmias, and their most serious con- A.V., N.N., L.V., B.R., and I.B. prepared figures; A.V., J.T., N.N.,
sequences, sudden cardiac death and stroke, improved E.P., B.R., and I.B. drafted manuscript; A.V., J.T., N.N., L.V., E.P.,

1136 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

B.R., and I.B. edited and revised manuscript; A.V., J.T., N.N., 17. Nguyen TP, Qu Z, Weiss JN. Cardiac fibrosis and arrhythmogene-
L.V., E.P., B.R., and I.B. approved final version of manuscript; sis: the road to repair is paved with perils. J Mol Cell Cardiol 70:
83–91, 2014. doi:10.1016/j.yjmcc.2013.10.018.

18. Varro A, Baczko I. Possible mechanisms of sudden cardiac death in


REFERENCES top athletes: a basic cardiac electrophysiological point of view.
Pflugers Arch 460: 31–40, 2010. doi:10.1007/s00424-010-0798-0.

1. Hoffman BF, Cranefield PF. Electrophysiology of the Heart. New 19. Mines GR. Further experiments on the action of the vagus on the
York: McGraw-Hill, 1960, p. 323. electrogram of the frog's heart. J Physiol 47: 419–430, 1914.
doi:10.1113/jphysiol.1914.sp001634.
2. Bers DM. Cardiac excitation-contraction coupling. Nature 415: 198–
205, 2002. doi:10.1038/415198a. 20. Coronel R, Wilms-Schopman FJ, Opthof T, Janse MJ. Dispersion of
repolarization and arrhythmogenesis. Heart Rhythm 6: 537–543,
3. Bartos DC, Grandi E, Ripplinger CM. Ion channels in the heart. 2009. doi:10.1016/j.hrthm.2009.01.013.
Compr Physiol 5: 1423–1464, 2015. doi:10.1002/cphy.c140069.
21. Opthof T, Janse MJ, Meijborg VM, Cinca J, Rosen MR, Coronel R.
4. Carmeliet E. Cardiac ionic currents and acute ischemia: from chan- Dispersion in ventricular repolarization in the human, canine and
nels to arrhythmias. Physiol Rev 79: 917–1017, 1999. doi:10.1152/ porcine heart. Prog Biophys Mol Biol 120: 222–235, 2016. doi:10.
physrev.1999.79.3.917. 1016/j.pbiomolbio.2016.01.007.
5. Schmitt N, Grunnet M, Olesen SP. Cardiac potassium channel sub- 22. Kline J, Costantini O. Inherited cardiac arrhythmias and channelo-
types: new roles in repolarization and arrhythmia. Physiol Rev 94: pathies. Med Clin North Am 103: 809–820, 2019. doi:10.1016/j.
609–653, 2014. doi:10.1152/physrev.00022.2013. mcna.2019.05.001.
6. Benjamin EJ, Wolf PA, D’Agostino RB, Silbershatz H, Kannel WB,
23. Skibsbye L, Ravens U. Mechanism of proarrhythmic effects of po-
Levy D. Impact of atrial fibrillation on the risk of death: the
tassium channel blockers. Card Electrophysiol Clin 8: 395–410,
Framingham Heart Study. Circulation 98: 946–952, 1998. doi:
2016. doi:10.1016/j.ccep.2016.02.004.
10.1161/01.cir.98.10.946.
24. Aiba T, Shimizu W, Hidaka I, Uemura K, Noda T, Zheng C, Kamiya
7. Nattel S, Maguy A, Le Bouter S, Yeh YH. Arrhythmogenic ion-chan-
A, Inagaki M, Sugimachi M, Sunagawa K. Cellular basis for trigger
nel remodeling in the heart: heart failure, myocardial infarction, and
and maintenance of ventricular fibrillation in the Brugada syndrome
atrial fibrillation. Physiol Rev 87: 425–456, 2007. doi:10.1152/
model: high-resolution optical mapping study. J Am Coll Cardiol
physrev.00014.2006.
47: 2074–2085, 2006. doi:10.1016/j.jacc.2005.12.064.
8. Temple IP, Inada S, Dobrzynski H, Boyett MR. Connexins and the
25. Akar FG, Rosenbaum DS. Transmural electrophysiological hetero-
atrioventricular node. Heart Rhythm 10: 297–304, 2013. doi:10.
geneities underlying arrhythmogenesis in heart failure. Circ Res
1016/j.hrthm.2012.10.020.
93: 638–645, 2003. doi:10.1161/01.RES.0000092248.59479.AE.
9. Attwell D, Cohen I, Eisner D, Ohba M, Ojeda C. The steady state
26. Antzelevitch C, Burashnikov A. Overview of basic mechanisms of
TTX-sensitive (“window”) sodium current in cardiac Purkinje fibres.
cardiac arrhythmia. Card Electrophysiol Clin 3: 23–45, 2011.
Pflugers Arch 379: 137–142, 1979. doi:10.1007/BF00586939.
doi:10.1016/j.ccep.2010.10.012.
10. January CT, Riddle JM. Early afterdepolarizations: mechanism of
27. Coppini R, Ferrantini C, Mugelli A, Poggesi C, Cerbai E. Altered
induction and block. A role for L-type Ca21 current. Circ Res 64:
Ca21 and Na1 homeostasis in human hypertrophic cardiomyopa-
977–990, 1989. doi:10.1161/01.res.64.5.977.
thy: implications for arrhythmogenesis. Front Physiol 9: 1391, 2018.
11. Banyasz T, Horvath B, Jian Z, Izu LT, Chen-Izu Y. Profile of L-type doi:10.3389/fphys.2018.01391.
Ca21 current and Na1/Ca21 exchange current during cardiac
28. de Bakker JM, van Capelle FJ, Janse MJ, Wilde AA, Coronel R,
action potential in ventricular myocytes. Heart Rhythm 9: 134–142,
Becker AE, Dingemans KP, van Hemel NM, Hauer RN. Reentry as a
2012. doi:10.1016/j.hrthm.2011.08.029.
cause of ventricular tachycardia in patients with chronic ischemic
12. Carmeliet E. Slow inactivation of the sodium current in rabbit car- heart disease: electrophysiologic and anatomic correlation.
diac Purkinje fibres. Pflugers Arch 408: 18–26, 1987. doi:10.1007/ Circulation 77: 589–606, 1988. doi:10.1161/01.cir.77.3.589.
BF00581835.
29. Harada M, Van Wagoner DR, Nattel S. Role of inflammation in atrial
13. Gintant GA, Datyner NB, Cohen IS. Slow inactivation of a tetrodo- fibrillation pathophysiology and management. Circ J 79: 495–502,
toxin-sensitive current in canine cardiac Purkinje fibers. Biophys J 2015. doi:10.1253/circj.CJ-15-0138.
45: 509–512, 1984. doi:10.1016/S0006-3495(84)84187-9.
30. Heijman J, Voigt N, Nattel S, Dobrev D. Cellular and molecular elec-
14. € ckner U. Calcium currents of isolated bovine ven-
Isenberg G, Klo trophysiology of atrial fibrillation initiation, maintenance, and pro-
tricular myocytes are fast and of large amplitude. Pflugers Arch gression. Circ Res 114: 1483–1499, 2014. doi:10.1161/CIRCRESAHA.
395: 30–41, 1982. doi:10.1007/BF00584965. 114.302226.

15. Kokubun S, Irisawa H. Effects of various intracellular Ca ion concen- 31. Janse MJ, Wit AL. Electrophysiological mechanisms of ventricular
trations on the calcium current of guinea-pig single ventricular cells. arrhythmias resulting from myocardial ischemia and infarction.
Jpn J Physiol 34: 599–611, 1984. doi:10.2170/jjphysiol.34.599. Physiol Rev 69: 1049–1169, 1989. doi:10.1152/physrev.1989.69.4.
1049.
16. Liu MB, de Lange E, Garfinkel A, Weiss JN, Qu Z. Delayed afterde-
polarizations generate both triggers and a vulnerable substrate 32. Lazzara R, Scherlag BJ. Electrophysiologic basis for arrhythmias in
promoting reentry in cardiac tissue. Heart Rhythm 12: 2115–2124, ischemic heart disease. Am J Cardiol 53: 1B–7B, 1984. doi:10.1016/
2015. doi:10.1016/j.hrthm.2015.06.019. 0002-9149(84)90493-4.

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1137


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

33. Park S, Nguyen NB, Pezhouman A, Ardehali R. Cardiac fibrosis: methods. Am J Heart Circ Physiol 270: H2157–H2168, 1996. doi:
potential therapeutic targets. Transl Res 209: 121–137, 2019. doi:10. 10.1152/ajpheart.1996.270.6.H2157.
1016/j.trsl.2019.03.001.
50. Pueyo E, Corrias A, Virág L, Jost N, Sze l T, Varro
 A, Szentandrássy
34. Schotten U, Verheule S, Kirchhof P, Goette A. Pathophysiological N, Nánási PP, Burrage K, Rodríguez B. A multiscale investigation of
mechanisms of atrial fibrillation: a translational appraisal. Physiol repolarization variability and its role in cardiac arrhythmogenesis.
Rev 91: 265–325, 2011. doi:10.1152/physrev.00031.2009. Biophys J 101: 2892–2902, 2011. doi:10.1016/j.bpj.2011.09.060.

35. Attwell D, Cohen I, Eisner DA. The effects of heart rate on the 51. Sharp GH, Joyner RW. Simulated propagation of cardiac action
action potential of guinea-pig and human ventricular muscle. J potentials. Biophys J 31: 403–423, 1980. doi:10.1016/S0006-3495
Physiol 313: 439–461, 1981. doi:10.1113/jphysiol.1981.sp013675. (80)85068-5.

36. Gadsby DC. Influence of Na/K pump current on action potentials in 52. Morad M, Salama G. Optical probes of membrane potential in heart
Purkinje fibers. Adv Myocardiol 5: 279–294, 1985. doi:10.1007/ muscle. J Physiol 292: 267–295, 1979. doi:10.1113/jphysiol.1979.
978-1-4757-1287-2_22. sp012850.

37. Gao J, Wang W, Cohen IS, Mathias RT. Transmural gradients in Na/ 53. Salama G, Lombardi R, Elson J. Maps of optical action potentials
K pump activity and [Na1]i in canine ventricle. Biophys J 89: 1700– and NADH fluorescence in intact working hearts. Am J Physiol
1709, 2005. doi:10.1529/biophysj.105.062406. Heart Circ Physiol 252: H384–H394, 1987. doi:10.1152/ajpheart.
1987.252.2.H384.
38. Lee JH. The Na/K pump, resting potential and selective permeabil-
ity in canine Purkinje fibres at physiologic and room temperatures. 54. Berenfeld O, Efimov I. Optical mapping. Card Electrophysiol Clin
Experientia 52: 657–660, 1996. doi:10.1007/BF01925568. 11: 495–510, 2019. doi:10.1016/j.ccep.2019.04.004.

39. Shaw RM, Rudy Y. Electrophysiologic effects of acute myocardial 55. Jaimes R 3rd, Walton RD, Pasdois P, Bernus O, Efimov IR, Kay MW.
ischemia: a theoretical study of altered cell excitability and action A technical review of optical mapping of intracellular calcium within
potential duration. Cardiovasc Res 35: 256–272, 1997. doi:10.1016/ myocardial tissue. Am J Physiol Heart Circ Physiol 310: H1388–
s0008-6363(97)00093-x. H1401, 2016. doi:10.1152/ajpheart.00665.2015.

56. Bishop MJ, Gavaghan DJ, Trayanova NA, Rodriguez B. Photon


40. Clauss S, Bleyer C, Sch€uttler D, Tomsits P, Renner S, Klymiuk N,
€a€b S. Animal models of arrhythmia: scattering effects in optical mapping of propagation and arrhythmo-
Wakili R, Massberg S, Wolf E, Ka
genesis in the heart. J Electrocardiol 40: S75–S80, 2007. doi:10.
classic electrophysiology to genetically modified large animals. Nat
1016/j.jelectrocard.2007.06.020.
Rev Cardiol 16: 457–475, 2019. doi:10.1038/s41569-019-0179-0.
57. Fedorov VV, Lozinsky IT, Sosunov EA, Anyukhovsky EP, Rosen MR,
41. Coraboeuf E, Weidmann S. Potentiels d’action du muscle cardia-
Balke CW, Efimov IR. Application of blebbistatin as an excitation-con-
que obtenus a l’aide de microelectrodes intracellilaires. Pre
 sence
traction uncoupler for electrophysiologic study of rat and rabbit hearts.
d’une inversion de potentiel. C R Soc Biol Paris 143: 1360–1361,
Heart Rhythm 4: 619–626, 2007. doi:10.1016/j.hrthm.2006.12.047.
1949.
58. Kanlop N, Sakai T. Optical mapping study of blebbistatin-induced
42. Coraboeuf E, Boistel J. [Study with the aid of intracellular micro-
chaotic electrical activities in isolated rat atrium preparations. J
electrodes of the action of a cardiotonic: 2-methyl-6-amino-2-hep-
Physiol Sci 60: 109–117, 2010. doi:10.1007/s12576-009-0074-2.
tanol on nodal tissue of dog heart]. C R Seances Soc Biol Fil 147:
774–779, 1953. 59. Brack KE, Narang R, Winter J, Ng GA. The mechanical uncoupler
blebbistatin is associated with significant electrophysiological
43. Franz MR. Method and theory of monophasic action potential re- effects in the isolated rabbit heart. Exp Physiol 98: 1009–1027,
cording. Prog Cardiovasc Dis 33: 347–368, 1991. doi:10.1016/0033- 2013. doi:10.1113/expphysiol.2012.069369.
0620(91)90002-4.
60. Banyasz T, Horvath B, Jian Z, Izu LT, Chen-Izu Y. Sequential dissec-
44. Harper RW, Olsson SB, Varnauskas E. Effect of mexiletine on tion of multiple ionic currents in single cardiac myocytes under
monophasic action potentials recorded from the right ventricle in action potential-clamp. J Mol Cell Cardiol 50: 578–581, 2011.
man. Cardiovasc Res 13: 303–310, 1979. doi:10.1093/cvr/13.6.303. doi:10.1016/j.yjmcc.2010.12.020.
45. Franz MR. Long-term recording of monophasic action potentials 61. Reuter H. Slow inactivation of currents in cardiac Purkinje fibres. J
from human endocardium. Am J Cardiol 51: 1629–1634, 1983. Physiol 197: 233–253, 1968. doi:10.1113/jphysiol.1968.sp008557.
doi:10.1016/0002-9149(83)90199-6.
62. Noble D. The Initiation of the Heartbeat. Oxford: Oxford University
46. Carmeliet E. From Bernstein’s rheotome to Neher-Sakmann’s patch Press, 1980.
electrode. The action potential. Physiol Rep 7: e13861, 2019.
doi:10.14814/phy2.13861. 63. Dhamoon AS, Pandit SV, Sarmast F, Parisian KR, Guha P, Li Y,
Bagwe S, Taffet SM, Anumonwo JM. Unique Kir2.x properties
47. Hamill OP, Marty A, Neher E, Sakmann B, Sigworth FJ. Improved determine regional and species differences in the cardiac inward
patch-clamp techniques for high-resolution current recording from rectifier K1 current. Circ Res 94: 1332–1339, 2004. doi:10.1161/01.
cells and cell-free membrane patches. Pflugers Arch 391: 85–100, RES.0000128408.66946.67.
1981. doi:10.1007/BF00656997.
64. Gurney A, Manoury B. Two-pore potassium channels in the cardio-
48. Neher E, Sakmann B. Single-channel currents recorded from mem- vascular system. Eur Biophys J 38: 305–318, 2009. doi:10.1007/
brane of denervated frog muscle fibres. Nature 260: 799–802, s00249-008-0326-8.
1976. doi:10.1038/260799a0.
65. Jones EM, Roti EC, Wang J, Delfosse SA, Robertson GA. Cardiac
49. Yue L, Feng J, Li GR, Nattel S. Transient outward and delayed recti- IKr channels minimally comprise hERG 1a and 1b subunits. J Biol
fier currents in canine atrium: properties and role of isolation Chem 279: 44690–44694, 2004. doi:10.1074/jbc.M408344200.

1138 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

66. Robertson GA, Jones EM, Wang J. Gating and assembly of hetero- 82. Coraboeuf E, Deroubaix E, Coulombe A. Effect of tetrodotoxin on
meric hERG1a/1b channels underlying IKr in the heart. Novartis action potentials of the conducting system in the dog heart. Am J
Found Symp 266: 4–15, 2005. Physiol Heart Circ Physiol 236: H561–H567, 1979. doi:10.1152/
ajpheart.1979.236.4.H561.
67. Bendahhou S, Marionneau C, Haurogne K, Larroque MM, Derand
R, Szuts V, Escande D, Demolombe S, Barhanin J. In vitro molecular 83. Di Stolfo G, Palumbo P, Castellana S, Mastroianno S, Biagini T,
interactions and distribution of KCNE family with KCNQ1 in the Palumbo O, Leone MP, De Luca G, Potenza DR, Mazza T, Russo
human heart. Cardiovasc Res 67: 529–538, 2005. doi:10.1016/j. AA, Carella M. Sudden cardiac death in J wave syndrome with
cardiores.2005.02.014. short QT associated to a novel mutation in Nav 1.8 coding gene
SCN10A: first case report for a possible pharmacogenomic role. J
68. Lundquist AL, Manderfield LJ, Vanoye CG, Rogers CS, Donahue Electrocardiol 51: 809–813, 2018. doi:10.1016/j.jelectrocard.2018.
BS, Chang PA, Drinkwater DC, Murray KT, George AL Jr. 06.005.
Expression of multiple KCNE genes in human heart may enable
variable modulation of IKs. J Mol Cell Cardiol 38: 277–287, 2005. 84. Felipe A, Knittle TJ, Doyle KL, Tamkun MM. Primary structure and
doi:10.1016/j.yjmcc.2004.11.012. differential expression during development and pregnancy of a
novel voltage-gated sodium channel in the mouse. J Biol Chem
69. Fozzard HA, January CT, Makielski JC. New studies of the excita- 269: 30125–30131, 1994. doi:10.1016/S0021-9258(18)43786-6.
tory sodium currents in heart muscle. Circ Res 56: 475–485, 1985.
doi:10.1161/01.res.56.4.475. 85. Gaborit N, Le Bouter S, Szuts V, Varro A, Escande D, Nattel S,
Demolombe S. Regional and tissue specific transcript signatures of
70. Rook MB, Evers MM, Vos MA, Bierhuizen MF. Biology of cardiac so- ion channel genes in the non-diseased human heart. J Physiol
dium channel Nav1.5 expression. Cardiovasc Res 93: 12–23, 2012. 582: 675–693, 2007. doi:10.1113/jphysiol.2006.126714.
doi:10.1093/cvr/cvr252.
86. Mishra S, Reznikov V, Maltsev VA, Undrovinas NA, Sabbah HN,
71. Jiang D, Shi H, Tonggu L, Gamal El-Din TM, Lenaeus MJ, Zhao Y, Undrovinas A. Contribution of sodium channel neuronal isoform
Yoshioka C, Zheng N, Catterall WA. Structure of the cardiac sodium Nav1.1 to late sodium current in ventricular myocytes from failing
channel. Cell 180: 122–134.e10, 2020. doi:10.1016/j.cell.2019.11.041. hearts. J Physiol 593: 1409–1427, 2015. doi:10.1113/jphysiol.2014.
278259.
€ cker D, Eickhorn R. Sodium current kinetics in intact rat
72. Antoni H, Bo
papillary muscle: measurements with the loose-patch-clamp tech- 87. Pabel S, Ahmad S, Tirilomis P, Stehle T, Mustroph J, Knierim M,
nique. J Physiol 406: 199–213, 1988. doi:10.1113/jphysiol.1988. € meke K,
Dybkova N, Bengel P, Holzamer A, Hilker M, Streckfuss-Bo
sp017376. Hasenfuss G, Maier LS, Sossalla S. Inhibition of NaV1.8 prevents
atrial arrhythmogenesis in human and mice. Basic Res Cardiol 115:
73. Brown AM, Lee KS, Powell T. Sodium current in single rat heart 20, 2020. doi:10.1007/s00395-020-0780-8.
muscle cells. J Physiol 318: 479–500, 1981. doi:10.1113/jphysiol.1981.
sp013879. 88. Savio-Galimberti E, Weeke P, Muhammad R, Blair M, Ansari S, Short
L, Atack TC, Kor K, Vanoye CG, Olesen MS, LuCamp, Yang T,
74. Fozzard HA, Friedlander IR, Hanck DA, January CT, Makielski George AL Jr, Roden DM, Darbar D. SCN10A/Nav1.8 modulation of
JC, Sheets MF. Sodium currents in single cardiac Purkinje cells. peak and late sodium currents in patients with early onset atrial fi-
J Am Coll Cardiol 8: 79A–85A, 1986. doi:10.1016/s0735-1097 brillation. Cardiovasc Res 104: 355–363, 2014. doi:10.1093/cvr/
(86)80033-x. cvu170.

75. Morad M, Tung L. Ionic events responsible for the cardiac resting 89. Biet M, Barajas-Martínez H, Ton AT, Delabre JF, Morin N, Dumaine
and action potential. Am J Cardiol 49: 584–594, 1982. doi:10.1016/ R. About half of the late sodium current in cardiac myocytes from
s0002-9149(82)80016-7. dog ventricle is due to non-cardiac-type Na1 channels. J Mol Cell
Cardiol 53: 593–598, 2012. doi:10.1016/j.yjmcc.2012.06.012.
76. Reuter H. Properties of two inward membrane currents in the heart.
Annu Rev Physiol 41: 413–424, 1979. doi:10.1146/annurev.ph.41. 90. Du Y, Huang X, Wang T, Han K, Zhang J, Xi Y, Wu G, Ma A.
030179.002213. Downregulation of neuronal sodium channel subunits Nav1.1 and
Nav1.6 in the sinoatrial node from volume-overloaded heart failure
77. Ten Eick RE, Baumgarten CM, Singer DH. Ventricular dysrhythmia: rat. Pflugers Arch 454: 451–459, 2007. doi:10.1007/s00424-007-
membrane basis or of currents, channels, gates, and cables. Prog 0216-4.
Cardiovasc Dis 24: 157–188, 1981. doi:10.1016/0033-0620(81)
90003-7. 91. Ahmad S, Tirilomis P, Pabel S, Dybkova N, Hartmann N, Molina CE,
Tirilomis T, Kutschka I, Frey N, Maier LS, Hasenfuss G, Streckfuss-
78. Bocchi L, Vassalle M. Characterization of the slowly inactivating so- € meke K, Sossalla S. The functional consequences of sodium
Bo
dium current INa2 in canine cardiac single Purkinje cells. Exp channel NaV1.8 in human left ventricular hypertrophy. ESC Heart
Physiol 93: 347–361, 2008. doi:10.1113/expphysiol.2007.040881. Fail 6: 154–163, 2019. doi:10.1002/ehf2.12378.

79. Maltsev VA, Undrovinas AI. A multi-modal composition of the late 92. Aguilar-Bryan L, Nichols CG, Wechsler SW, Clement JP 4th, Boyd
Na1 current in human ventricular cardiomyocytes. Cardiovasc Res AE 3rd, González G, Herrera-Sosa H, Nguy K, Bryan J, Nelson DA.
69: 116–127, 2006. doi:10.1016/j.cardiores.2005.08.015. Cloning of the beta cell high-affinity sulfonylurea receptor: a regula-
tor of insulin secretion. Science 268: 423–426, 1995. doi:10.1126/
80. Kiyosue T, Arita M. Late sodium current and its contribution to science.7716547.
action potential configuration in guinea pig ventricular myocytes.
Circ Res 64: 389–397, 1989. doi:10.1161/01.res.64.2.389. 93. Maier SK, Westenbroek RE, Schenkman KA, Feigl EO, Scheuer T,
Catterall WA. An unexpected role for brain-type sodium channels
81. Zaza A, Belardinelli L, Shryock JC. Pathophysiology and pharmacol- in coupling of cell surface depolarization to contraction in the heart.
ogy of the cardiac “late sodium current”. Pharmacol Ther 119: 326– Proc Natl Acad Sci USA 99: 4073–4078, 2002. doi:10.1073/
339, 2008. doi:10.1016/j.pharmthera.2008.06.001. pnas.261705699.

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1139


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

94. Rivaud MR, Delmar M, Remme CA. Heritable arrhythmia syndromes 108. Lei M, Wu L, Terrar DA, Huang CL. Modernized classification of car-
associated with abnormal cardiac sodium channel function: ionic diac antiarrhythmic drugs. Circulation 138: 1879–1896, 2018. doi:
and non-ionic mechanisms. Cardiovasc Res 116: 1557–1570, 2020. 10.1161/CIRCULATIONAHA.118.035455.
doi:10.1093/cvr/cvaa082.
109. Roden DM. Pharmacology and toxicology of Nav1.5-class 1 anti-
95. Leo-Macias A, Agullo-Pascual E, Sanchez-Alonso JL, Keegan S, Lin arrhythmic drugs. Card Electrophysiol Clin 6: 695–704, 2014.
X, Arcos T, Feng-Xia L, Korchev YE, Gorelik J, Fenyo € D, doi:10.1016/j.ccep.2014.07.003.
Rothenberg E, Rothenberg E, Delmar M. Nanoscale visualization of
functional adhesion/excitability nodes at the intercalated disc. Nat 110. Coppini R, Santini L, Olivotto I, Ackerman MJ, Cerbai E.
Commun 7: 10342, 2016. doi:10.1038/ncomms10342. Abnormalities in sodium current and calcium homoeostasis as
drivers of arrhythmogenesis in hypertrophic cardiomyopathy.
96. Rougier JS, Essers MC, Gillet L, Guichard S, Sonntag S, Shmerling Cardiovasc Res 116: 1585–1599, 2020. doi:10.1093/cvr/cvaa124.
D, Abriel H. A distinct pool of Nav1.5 channels at the lateral mem-
brane of murine ventricular cardiomyocytes. Front Physiol 10: 834, 111. Rajamani S, Liu G, El-Bizri N, Guo D, Li C, Chen XL, Kahlig KM,
2019. doi:10.3389/fphys.2019.00834. Mollova N, Elzein E, Zablocki J, Belardinelli L. The novel late Na1
current inhibitor, GS-6615 (eleclazine) and its anti-arrhythmic effects
97. Lin X, Liu N, Lu J, Zhang J, Anumonwo JM, Isom LL, Fishman GI, in rabbit isolated heart preparations. Br J Pharmacol 173: 3088–
Delmar M. Subcellular heterogeneity of sodium current properties 3098, 2016. doi:10.1111/bph.13563.
in adult cardiac ventricular myocytes. Heart Rhythm 8: 1923–1930,
2011. doi:10.1016/j.hrthm.2011.07.016. 112. Le Grand B, Coulombe A, John GW. Late sodium current inhibition in
human isolated cardiomyocytes by R 56865. J Cardiovasc
98. Rivaud MR, Agullo-Pascual E, Lin X, Leo-Macias A, Zhang M, Pharmacol 31: 800–804, 1998. doi:10.1097/00005344-199805000-
Rothenberg E, Bezzina CR, Delmar M, Remme CA. Sodium channel
00021.
remodeling in subcellular microdomains of murine failing cardiomy-
ocytes. J Am Heart Assoc 6: e007622, 2017. doi:10.1161/JAHA.117. 113. Wu M, Tran PN, Sheng J, Randolph AL, Wu WW. Drug potency on
007622. inhibiting late Na1 current is sensitive to gating modifier and cur-
rent region where drug effects were measured. J Pharmacol
99. Johnson CN. Calcium modulation of cardiac sodium channels. J
Toxicol Methods 100: 106605, 2019. doi:10.1016/j.vascn.2019.
Physiol 598: 2835–2846, 2020. doi:10.1113/JP277553.
106605.
100. Tan HL, Kupershmidt S, Zhang R, Stepanovic S, Roden DM, Wilde
114. Bohnen MS, Peng G, Robey SH, Terrenoire C, Iyer V, Sampson KJ,
AA, Anderson ME, Balser JR. A calcium sensor in the sodium chan-
Kass RS. Molecular pathophysiology of congenital long QT syn-
nel modulates cardiac excitability. Nature 415: 442–447, 2002.
drome. Physiol Rev 97: 89–134, 2017. doi:10.1152/physrev.00008.
doi:10.1038/415442a.
2016.
101. Johnson EK, Springer SJ, Wang W, Dranoff EJ, Zhang Y, Kanter EM,
Yamada KA, Nerbonne JM. Differential expression and remodeling 115. Giudicessi JR, Wilde AA, Ackerman MJ. The genetic architecture of
of transient outward potassium currents in human left ventricles. long QT syndrome: a critical reappraisal. Trends Cardiovasc Med
Circ Arrhythm Electrophysiol 11: e005914, 2018. doi:10.1161/ 28: 453–464, 2018. doi:10.1016/j.tcm.2018.03.003.
CIRCEP.117.005914.
116. Gourraud JB, Barc J, Thollet A, Le Marec H, Probst V. Brugada syn-
102. Gabelli SB, Boto A, Kuhns VH, Bianchet MA, Farinelli F, Aripirala S, drome: diagnosis, risk stratification and management. Arch
Yoder J, Jakoncic J, Tomaselli GF, Amzel LM. Regulation of the Cardiovasc Dis 110: 188–195, 2017. doi:10.1016/j.acvd.2016.09.009.
NaV1.5 cytoplasmic domain by calmodulin. Nat Commun 5: 5126,
117. Skinner JR, Winbo A, Abrams D, Vohra J, Wilde AA. Channelopathies
2014. doi:10.1038/ncomms6126.
that lead to sudden cardiac death: clinical and genetic aspects. Heart
103. Johnson C, Forsythe L, Somauroo J, Papadakis M, George K, Lung Circ 28: 22–30, 2019. doi:10.1016/j.hlc.2018.09.007.
Oxborough D. Cardiac structure and function in elite Native
118. Boyden PA, Dun W, Barbhaiya C, Ter Keurs HE. 2APB- and JTV519
Hawaiian and Pacific Islander Rugby Football League athletes: an
(K201)-sensitive micro Ca21 waves in arrhythmogenic Purkinje cells
exploratory study. Int J Cardiovasc Imaging 34: 725–734, 2018.
doi:10.1007/s10554-017-1285-x. that survive in infarcted canine heart. Heart Rhythm 1: 218–226,
2004. doi:10.1016/j.hrthm.2004.03.068.
104. Wang C, Chung BC, Yan H, Wang HG, Lee SY, Pitt GS. Structural
analyses of Ca21/CaM interaction with NaV channel C-termini 119. Benson DW, Wang DW, Dyment M, Knilans TK, Fish FA, Strieper
reveal mechanisms of calcium-dependent regulation. Nat MJ, Rhodes TH, George AL Jr. Congenital sick sinus syndrome
Commun 5: 4896, 2014. doi:10.1038/ncomms5896. caused by recessive mutations in the cardiac sodium channel gene
(SCN5A). J Clin Invest 112: 1019–1028, 2003. doi:10.1172/JCI18062.
105. Maltsev VA, Reznikov V, Undrovinas NA, Sabbah HN, Undrovinas
A. Modulation of late sodium current by Ca21, calmodulin, and 120. Schott JJ, Alshinawi C, Kyndt F, Probst V, Hoorntje TM, Hulsbeek
CaMKII in normal and failing dog cardiomyocytes: similarities and M, Wilde AA, Escande D, Mannens MM, Le Marec H. Cardiac con-
differences. Am J Physiol Heart Circ Physiol 294: H1597–H1608, duction defects associate with mutations in SCN5A. Nat Genet 23:
2008. doi:10.1152/ajpheart.00484.2007. 20–21, 1999. doi:10.1038/12618.

106. Wagner S, Dybkova N, Rasenack EC, Jacobshagen C, Fabritz L, 121. Smits JP, Koopmann TT, Wilders R, Veldkamp MW, Opthof T,
Kirchhof P, Maier SK, Zhang T, Hasenfuss G, Brown JH, Bers DM, Bhuiyan ZA, Mannens MM, Balser JR, Tan HL, Bezzina CR, Wilde
Maier LS. Ca21/calmodulin-dependent protein kinase II regulates AA. A mutation in the human cardiac sodium channel (E161K) con-
cardiac Na1 channels. J Clin Invest 116: 3127–3138, 2006. tributes to sick sinus syndrome, conduction disease and Brugada
doi:10.1172/JCI26620. syndrome in two families. J Mol Cell Cardiol 38: 969–981, 2005.
doi:10.1016/j.yjmcc.2005.02.024.
107. Anderson ME, Brown JH, Bers DM. CaMKII in myocardial hypertro-
phy and heart failure. J Mol Cell Cardiol 51: 468–473, 2011. 122. Wang Q, Shen J, Li Z, Timothy K, Vincent GM, Priori SG, Schwartz
doi:10.1016/j.yjmcc.2011.01.012. PJ, Keating MT. Cardiac sodium channel mutations in patients with

1140 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

long QT syndrome, an inherited cardiac arrhythmia. Hum Mol myocytes of human left ventricle. Circulation 93: 168–177, 1996.
Genet 4: 1603–1607, 1995. doi:10.1093/hmg/4.9.1603. doi:10.1161/01.cir.93.1.168.doi:10.1161/01.CIR.93.1.168.

123. Li Q, Huang H, Liu G, Lam K, Rutberg J, Green MS, Birnie DH, 137. Patel SP, Campbell DL. Transient outward potassium current, ‘Ito’,
Lemery R, Chahine M, Gollob MH. Gain-of-function mutation of phenotypes in the mammalian left ventricle: underlying molecular,
Nav1.5 in atrial fibrillation enhances cellular excitability and lowers cellular and biophysical mechanisms. J Physiol 569: 7–39, 2005.
the threshold for action potential firing. Biochem Biophys Res doi:10.1113/jphysiol.2005.086223.
Commun 380: 132–137, 2009. doi:10.1016/j.bbrc.2009.01.052.
138. Wang Z, Feng J, Shi H, Pond A, Nerbonne JM, Nattel S. Potential
124. Makiyama T, Akao M, Shizuta S, Doi T, Nishiyama K, Oka Y, Ohno molecular basis of different physiological properties of the transient
S, Nishio Y, Tsuji K, Itoh H, Kimura T, Kita T, Horie M. A novel outward K1 current in rabbit and human atrial myocytes. Circ Res
SCN5A gain-of-function mutation M1875T associated with familial 84: 551–561, 1999. doi:10.1161/01.res.84.5.551.
atrial fibrillation. J Am Coll Cardiol 52: 1326–1334, 2008. doi:10.
139. Ordo€ g B, Brutyo  E, Puskás LG, Papp JG, Varro
 A, Szabad J,
1016/j.jacc.2008.07.013.
Boldogkoi Z. Gene expression profiling of human cardiac potas-
125. Remme CA, Wilde AA. Late sodium current inhibition in acquired sium and sodium channels. Int J Cardiol 111: 386–393, 2006.
and inherited ventricular (dys)function and arrhythmias. Cardiovasc doi:10.1016/j.ijcard.2005.07.063.
Drugs Ther 27: 91–101, 2013. doi:10.1007/s10557-012-6433-x.
140. Virág L, Jost N, Papp R, Koncz I, Kristo  f A, Kohajda Z, Harmati G,
126. Undrovinas AI, Maltsev VA, Kyle JW, Silverman N, Sabbah HN. Carbonell-Pascual B, Ferrero JM Jr, Papp JG, Nanási PP, Varro  A.
Gating of the late Na1 channel in normal and failing human myocar- Analysis of the contribution of Ito to repolarization in canine ventric-
dium. J Mol Cell Cardiol 34: 1477–1489, 2002. doi:10.1006/jmcc. ular myocardium. Br J Pharmacol 164: 93–105, 2011. doi:10.1111/
2002.2100. j.1476-5381.2011.01331.x.

127. Undrovinas AI, Maltsev VA, Sabbah HN. Repolarization abnormal- 141. Han W, Wang Z, Nattel S. A comparison of transient outward cur-
ities in cardiomyocytes of dogs with chronic heart failure: role of rents in canine cardiac Purkinje cells and ventricular myocytes. Am
sustained inward current. Cell Mol Life Sci 55: 494–505, 1999. J Physiol Heart Circ Physiol 279: H466–H474, 2000. doi:10.1152/
doi:10.1007/s000180050306. ajpheart.2000.279.2.H466.

142. Surawicz B. Role of potassium channels in cycle length dependent


128. Antzelevitch C, Fish J. Electrical heterogeneity within the ventricu-
regulation of action potential duration in mammalian cardiac
lar wall. Basic Res Cardiol 96: 517–527, 2001. doi:10.1007/s0039
Purkinje and ventricular muscle fibres. Cardiovasc Res 26: 1021–
50170002.
1029, 1992. doi:10.1093/cvr/26.11.1021.
129. Ferrier GR. Digitalis arrhythmias: role of oscillatory afterpotentials.
143. Chiamvimonvat N, Chen-Izu Y, Clancy CE, Deschenes I, Dobrev D,
Prog Cardiovasc Dis 19: 459–474, 1977. [ doi:10.1016/0033-0620
Heijman J, Izu L, Qu Z, Ripplinger CM, Vandenberg JI, Weiss JN,
(77)90010-X.
Koren G, Banyasz T, Grandi E, Sanguinetti MC, Bers DM, Nerbonne
130. Ferrier GR, Moe GK. Effect of calcium on acetylstrophanthidin- JM. Potassium currents in the heart: functional roles in repolariza-
induced transient depolarizations in canine Purkinje tissue. Circ tion, arrhythmia and therapeutics. J Physiol 595: 2229–2252,
Res 33: 508–515, 1973. doi:10.1161/01.res.33.5.508. 2017. doi:10.1113/JP272883.

131. Szabo B, Kovacs T, Lazzara R. Role of calcium loading in early after-  A, Lathrop DA, Hester SB, Nanási PP, Papp JG. Ionic currents
144. Varro
depolarizations generated by Cs1 in canine and guinea pig and action potentials in rabbit, rat, and guinea pig ventricular myo-
Purkinje fibers. J Cardiovasc Electrophysiol 6: 796–812, 1995. cytes. Basic Res Cardiol 88: 93–102, 1993. doi:10.1007/BF007
doi:10.1111/j.1540-8167.1995.tb00356.x. 98257.

132. Dehghani-Samani A, Madreseh-Ghahfarokhi S, Dehghani-Samani 145. Zicha S, Moss I, Allen B, Varro A, Papp J, Dumaine R, Antzelevich C,
A. Mutations of voltage-gated ionic channels and risk of severe car- Nattel S. Molecular basis of species-specific expression of repola-
diac arrhythmias. Acta Cardiol Sin 35: 99–110, 2019. doi:10.6515/ rizing K1 currents in the heart. Am J Physiol Heart Circ Physiol
285: H1641–H1649, 2003. doi:10.1152/ajpheart.00346.2003.
ACS.201903_35(2).20181028A.
146. Thomas SP, Bircher-Lehmann L, Thomas SA, Zhuang J, Saffitz JE,
133. Foeger NC, Norris AJ, Wren LM, Nerbonne JM. Augmentation of
ber AG. Synthetic strands of neonatal mouse cardiac myocytes:
Kle
Kv4.2-encoded currents by accessory dipeptidyl peptidase 6 and
structural and electrophysiological properties. Circ Res 87: 467–
10 subunits reflects selective cell surface Kv4.2 protein stabiliza-
473, 2000. doi:10.1161/01.RES.87.6.467.
tion. J Biol Chem 287: 9640–9650, 2012. doi:10.1074/jbc.M111.
324574. 147. Wang L, Duff HJ. Developmental changes in transient outward cur-
rent in mouse ventricle. Circ Res 81: 120–127, 1997. doi:10.1161/01.
134. Jimenez J, Rentschler SL. Transcriptional and epigenetic regulation
RES.81.1.120.
of cardiac electrophysiology. Pediatr Cardiol 40: 1325–1330, 2019.
doi:10.1007/s00246-019-02160-w. 148. Turnow K, Metzner K, Cotella D, Morales MJ, Schaefer M, Christ T,
Ravens U, Wettwer E, Kammerer S. Interaction of DPP10a with
135. Ka€a€b S, Dixon J, Duc J, Ashen D, Na
€bauer M, Beuckelmann DJ,
Kv4.3 channel complex results in a sustained current component of
Steinbeck G, McKinnon D, Tomaselli GF. Molecular basis of tran-
human transient outward current Ito. Basic Res Cardiol 110: 5,
sient outward potassium current downregulation in human heart
2015. doi:10.1007/s00395-014-0457-2.
failure: a decrease in Kv4.3 mRNA correlates with a reduction in
current density. Circulation 98: 1383–1393, 1998. doi:10.1161/01. 149. Wilde AA, Bezzina CR. Genetics of cardiac arrhythmias. Heart 91:
cir.98.14.1383. 1352–1358, 2005. doi:10.1136/hrt.2004.046334.

136. Na€bauer M, Beuckelmann DJ, Ûberfuhr P, Steinbeck G. Regional 150. Di Diego JM, Cordeiro JM, Goodrow RJ, Fish JM, Zygmunt AC,
differences in current density and rate-dependent properties of the rez GJ, Scornik FS, Antzelevitch C. Ionic and cellular basis for the
Pe
transient outward current in subepicardial and subendocardial predominance of the Brugada syndrome phenotype in males.

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1141


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

Circulation 106: 2004–2011, 2002. doi:10.1161/01.CIR.0000032002. Mohler PJ, Gyo €rke S, Carnes CA. Heart failure duration progres-
22105.7A. sively modulates the arrhythmia substrate through structural and
electrical remodeling. Life Sci 123: 61–71, 2015. doi:10.1016/j.
151. Gaborit N, Varro A, Le Bouter S, Szuts V, Escande D, Nattel S, lfs.2014.12.024.
Demolombe S. Gender-related differences in ion-channel and
transporter subunit expression in non-diseased human hearts. J 165. Na€bauer M, Beuckelmann DJ, Erdmann E. Characteristics of tran-
Mol Cell Cardiol 49: 639–646, 2010. doi:10.1016/j.yjmcc.2010. sient outward current in human ventricular myocytes from patients
06.005. with terminal heart failure. Circ Res 73: 386–394, 1993. doi:10.1161/
01.res.73.2.386.
152. Wang Z, Fermini B, Nattel S. Effects of flecainide, quinidine, and 4-
aminopyridine on transient outward and ultrarapid delayed rectifier 166. Sah R, Ramirez RJ, Backx PH. Modulation of Ca21 release in car-
currents in human atrial myocytes. J Pharmacol Exp Ther 272: diac myocytes by changes in repolarization rate: role of phase-1
184–196, 1995. action potential repolarization in excitation-contraction coupling.
Circ Res 90: 165–173, 2002. doi:10.1161/hh0202.103315.
153. Diochot S, Drici MD, Moinier D, Fink M, Lazdunski M. Effects of
phrixotoxins on the Kv4 family of potassium channels and implica- 167. van der Heyden MA, Wijnhoven TJ, Opthof T. Molecular aspects of
tions for the role of Ito1 in cardiac electrogenesis. Br J Pharmacol adrenergic modulation of the transient outward current.
126: 251–263, 1999. doi:10.1038/sj.bjp.0702283. Cardiovasc Res 71: 430–442, 2006. doi:10.1016/j.cardiores.2006.
154. Christ T, Wettwer E, Voigt N, Hála O, Radicke S, Matschke K, Varro 04.012.
A, Dobrev D, Ravens U. Pathology-specific effects of the IKur/Ito/IK, 168. Alonso H, Fernández-Ruocco J, Gallego M, Malagueta-Vieira LL,
ACh blocker AVE0118 on ion channels in human chronic atrial fibril- Rodríguez-de-Yurre A, Medei E, Casis O. Thyroid stimulating hor-
lation. Br J Pharmacol 154: 1619–1630, 2008. doi:10.1038/bjp.2008.
mone directly modulates cardiac electrical activity. J Mol Cell
209.
Cardiol 89: 280–286, 2015. doi:10.1016/j.yjmcc.2015.10.019.
155. Calloe K, Nof E, Jespersen T, Di Diego JM, Chlus N, Olesen SP,
169. Nishiyama A, Kambe F, Kamiya K, Seo H, Toyama J. Effects of thy-
Antzelevitch C, Cordeiro JM. Comparison of the effects of a transient
roid status on expression of voltage-gated potassium channels in
outward potassium channel activator on currents recorded from
rat left ventricle. Cardiovasc Res 40: 343–351, 1998. doi:10.1016/
atrial and ventricular cardiomyocytes. J Cardiovasc Electrophysiol
S0008-6363(98)00135-7.
22: 1057–1066, 2011. doi:10.1111/j.1540-8167.2011.02053.x.

€bauer M, Erdmann E. Alterations of K1 cur- 170. Reuter H. The dependence of slow inward current in Purkinje fibres
156. Beuckelmann DJ, Na
on the extracellular calcium-concentration. J Physiol 192: 479–492,
rents in isolated human ventricular myocytes from patients with ter-
1967. doi:10.1113/jphysiol.1967.sp008310.
minal heart failure. Circ Res 73: 379–385, 1993. doi:10.1161/01.
RES.73.2.379. 171. Hirano Y, Fozzard HA, January CT. Characteristics of L- and T-type
157. Han W, Chartier D, Li D, Nattel S. Ionic remodeling of cardiac Ca21 currents in canine cardiac Purkinje cells. Am J Physiol Heart
Purkinje cells by congestive heart failure. Circulation 104: 2095– Circ Physiol 256: H1478–H1492, 1989. doi:10.1152/ajpheart.1989.
2100, 2001. doi:10.1161/hc4201.097134. 256.5.H1478.

158. Han W, Zhang L, Schram G, Nattel S. Properties of potassium cur- 172. Mitra R, Morad M. Two types of calcium channels in guinea pig ven-
rents in Purkinje cells of failing human hearts. Am J Physiol Heart tricular myocytes. Proc Natl Acad Sci USA 83: 5340–5344, 1986.
Circ Physiol 283: H2495–H2503, 2002. doi:10.1152/ajpheart. doi:10.1073/pnas.83.14.5340.
00389.2002.
173. Hofmann F, Flockerzi V, Kahl S, Wegener JW. L-type CaV1.2 cal-
159. Zicha S, Xiao L, Stafford S, Cha TJ, Han W, Varro A, Nattel S. cium channels: from in vitro findings to in vivo function. Physiol Rev
Transmural expression of transient outward potassium current sub- 94: 303–326, 2014. doi:10.1152/physrev.00016.2013.
units in normal and failing canine and human hearts. J Physiol 561:
174. Catterall WA. Structure and function of voltage-sensitive ion chan-
735–748, 2004. doi:10.1113/jphysiol.2004.075861.
nels. Science 242: 50–61, 1988. doi:10.1126/science.2459775.
160. Coppini R, Ferrantini C, Yao L, Fan P, Del Lungo M, Stillitano F,
Sartiani L, Tosi B, Suffredini S, Tesi C, Yacoub M, Olivotto I, 175. Buraei Z, Yang J. The b subunit of voltage-gated Ca21 channels.
Belardinelli L, Poggesi C, Cerbai E, Mugelli A. Late sodium current Physiol Rev 90: 1461–1506, 2010. doi:10.1152/physrev.00057.
inhibition reverses electromechanical dysfunction in human hyper- 2009.
trophic cardiomyopathy. Circulation 127: 575–584, 2013.
176. Davies A, Hendrich J, Van Minh AT, Wratten J, Douglas L, Dolphin
doi:10.1161/CIRCULATIONAHA.112.134932.
AC. Functional biology of the alpha(2)delta subunits of voltage-
161. Keller KM, Howlett SE. Sex differences in the biology and pathol- gated calcium channels. Trends Pharmacol Sci 28: 220–228,
ogy of the aging heart. Can J Cardiol 32: 1065–1073, 2016. 2007. doi:10.1016/j.tips.2007.03.005.
doi:10.1016/j.cjca.2016.03.017.
177. Kang MG, Campbell KP. Gamma subunit of voltage-activated cal-
162. Regitz-Zagrosek V, Kararigas G. Mechanistic pathways of sex dif- cium channels. J Biol Chem 278: 21315–21318, 2003. doi:10.1074/
ferences in cardiovascular disease. Physiol Rev 97: 1–37, 2017. jbc.R300004200.
doi:10.1152/physrev.00021.2015.
178. Bers DM. Calcium cycling and signaling in cardiac myocytes. Annu
163. Cordeiro JM, Calloe K, Aschar-Sobbi R, Kim KH, Korogyi A, Rev Physiol 70: 23–49, 2008. doi:10.1146/annurev.physiol.70.
Occhipinti D, Backx PH, Panama BK. Physiological roles of the tran- 113006.100455.
sient outward current Ito in normal and diseased hearts. Front
Biosci (Schol Ed) 8: 143–159, 2016. doi:10.2741/s454. 179. Dibb KM, Eisner DA, Trafford AW. Regulation of systolic [Ca21]i and
cellular Ca21 flux balance in rat ventricular myocytes by SR Ca21,
164. Long VP 3rd, Bonilla IM, Vargas-Pinto P, Nishijima Y, Sridhar A, Li C, L-type Ca21 current and diastolic [Ca21]i. J Physiol 585: 579–592,
Mowrey K, Wright P, Velayutham M, Kumar S, Lee NY, Zweier JL, 2007. doi:10.1113/jphysiol.2007.141473.

1142 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

180. Mesirca P, Torrente AG, Mangoni ME. Functional role of voltage 195. Zhang S, Zhou Z, Gong Q, Makielski JC, January CT. Mechanism of
gated Ca21 channels in heart automaticity. Front Physiol 6: 19, block and identification of the verapamil binding domain to HERG
2015. doi:10.3389/fphys.2015.00019. potassium channels. Circ Res 84: 989–998, 1999. doi:10.1161/01.
res.84.9.989.
181. Adachi-Akahane S, Cleemann L, Morad M. Cross-signaling
between L-type Ca21 channels and ryanodine receptors in rat ven- 196. Koidl B, Miyawaki N, Tritthart HA. A novel benzothiazine Ca21
tricular myocytes. J Gen Physiol 108: 435–454, 1996. doi:10.1085/ channel antagonist, semotiadil, inhibits cardiac L-type Ca21 cur-
jgp.108.5.435. rents. Eur J Pharmacol 322: 243–247, 1997. doi:10.1016/s0014-
2999(96)00995-8.
182. Bechem M, Pott L. Evidence for Ca-mediated inactivation of ICa in
dialysed guinea-pig atrial cardioballs. Basic Res Cardiol 80: 101– 197. Hume JR. Comparative interactions of organic Ca11 channel
105, 1985. doi:10.1007/978-3-662-11041-6_20. antagonists with myocardial Ca11 and K1 channels. J Pharmacol
Exp Ther 234: 134–140, 1985.
183. Kass RS, Sanguinetti MC. Inactivation of calcium channel current in
the calf cardiac Purkinje fiber. Evidence for voltage- and calcium- 198. Morad M, Goldman YE, Trentham DR. Rapid photochemical inacti-
mediated mechanisms. J Gen Physiol 84: 705–726, 1984. vation of Ca21-antagonists shows that Ca21 entry directly activates
doi:10.1085/jgp.84.5.705. contraction in frog heart. Nature 304: 635–638, 1983. doi:10.1038/
304635a0.
184. Sham JS. Ca21 release-induced inactivation of Ca21 current in rat
ventricular myocytes: evidence for local Ca21 signalling. J Physiol 199. Artigas P, Ferreira G, Reyes N, Brum G, Pizarro G. Effects of the
500: 285–295, 1997. doi:10.1113/jphysiol.1997.sp022020. enantiomers of BayK 8644 on the charge movement of L-type Ca
channels in guinea-pig ventricular myocytes. J Membr Biol 193:
185. Tseng GN, Boyden PA. Multiple types of Ca21 currents in single ca- 215–227, 2003. doi:10.1007/s00232-003-2020-1.
nine Purkinje cells. Circ Res 65: 1735–1750, 1989. doi:10.1161/01.
200. Leprán I, Baczko  I, Varro
 A, Papp JG. ATP-sensitive potassium
res.65.6.1735.
channel modulators: both pinacidil and glibenclamide produce anti-
186. Ochi R, Trautwein W. The dependence of cardiac contraction on arrhythmic activity during acute myocardial infarction in conscious
depolarization and slow inward current. Pflugers Arch 323: 187– rats. J Pharmacol Exp Ther 277: 1215–1220, 1996.
203, 1971. doi:10.1007/BF00586383.
201. Martinez ME, Walton RD, Bayer JD, Haïssaguerre M, Vigmond EJ,
187. Isenberg G. Ca entry and contraction as studied in isolated bovine Hocini M, Bernus O. Role of the Purkinje-muscle junction on the
ventricular myocytes. Z Naturforsch C Biosci 37: 502–512, 1982. ventricular repolarization heterogeneity in the healthy and ischemic
doi:10.1515/znc-1982-5-623. ovine ventricular myocardium. Front Physiol 9: 718, 2018.
doi:10.3389/fphys.2018.00718.
188. Z€
uhlke RD, Pitt GS, Deisseroth K, Tsien RW, Reuter H. Calmodulin
supports both inactivation and facilitation of L-type calcium chan- 202. Hondeghem LM, Carlsson L, Duker G. Instability and triangulation
nels. Nature 399: 159–162, 1999. doi:10.1038/20200. of the action potential predict serious proarrhythmia, but action
potential duration prolongation is antiarrhythmic. Circulation 103:
189. Anderson ME, Braun AP, Schulman H, Premack BA. Multifunctional 2004–2013, 2001. doi:10.1161/01.CIR.103.15.2004.
Ca21/calmodulin-dependent protein kinase mediates Ca21-
203. Rigg L, Terrar DA. Possible role of calcium release from the sarco-
induced enhancement of the L-type Ca21 current in rabbit ventricu-
plasmic reticulum in pacemaking in guinea-pig sino-atrial node.
lar myocytes. Circ Res 75: 854–861, 1994. doi:10.1161/01.RES.
Exp Physiol 81: 877–880, 1996. doi:10.1113/expphysiol.1996.
75.5.854.
sp003983.
190. Yuan W, Bers DM. Ca-dependent facilitation of cardiac Ca current
204. Suenari K, Chen YC, Kao YH, Cheng CC, Lin YK, Chen YJ, Chen SA.
is due to Ca-calmodulin-dependent protein kinase. Am J Physiol
Discrepant electrophysiological characteristics and calcium home-
Heart Circ Physiol 267: H982–H993, 1994. doi:10.1152/ajpheart.
ostasis of left atrial anterior and posterior myocytes. Basic Res
1994.267.3.H982.
Cardiol 106: 65–74, 2011. doi:10.1007/s00395-010-0132-1.
191. Bers DM, Morotti S. Ca21 current facilitation is CaMKII-dependent 205. Liang B, Nissen JD, Laursen M, Wang X, Skibsbye L, Hearing MC,
and has arrhythmogenic consequences. Front Pharmacol 5: 144, Andersen MN, Rasmussen HB, Wickman K, Grunnet M, Olesen SP,
2014. doi:10.3389/fphar.2014.00144. Jespersen T. G-protein-coupled inward rectifier potassium current
192. Liu G, Papa A, Katchman AN, Zakharov SI, Roybal D, Hennessey contributes to ventricular repolarization. Cardiovasc Res 101: 175–
184, 2014. doi:10.1093/cvr/cvt240.
JA, Kushner J, Yang L, Chen BX, Kushnir A, Dangas K, Gygi SP, Pitt
GS, Colecraft HM, Ben-Johny M, Kalocsay M, Marx SO. Mechanism 206. Barrett CF, Tsien RW. The Timothy syndrome mutation differentially
of adrenergic CaV1.2 stimulation revealed by proximity proteomics. affects voltage- and calcium-dependent inactivation of CaV1.2 L-
Nature 577: 695–700, 2020. doi:10.1038/s41586-020-1947-z. type calcium channels. Proc Natl Acad Sci USA 105: 2157–2162,
2008. doi:10.1073/pnas.0710501105.
193. Rolf S, Haverkamp W, Borggrefe M, Musshoff U, Eckardt L,
Mergenthaler J, Snyders DJ, Pongs O, Speckmann EJ, Breithardt G, 207. Splawski I, Timothy KW, Sharpe LM, Decher N, Kumar P, Bloise R,
Madeja M. Effects of antiarrhythmic drugs on cloned cardiac volt- Napolitano C, Schwartz PJ, Joseph RM, Condouris K, Tager-
age-gated potassium channels expressed in Xenopus oocytes. Flusberg H, Priori SG, Sanguinetti MC, Keating MT. CaV1.2 calcium
Naunyn Schmiedebergs Arch Pharmacol 362: 22–31, 2000. channel dysfunction causes a multisystem disorder including ar-
doi:10.1007/s002100000257. rhythmia and autism. Cell 119: 19–31, 2004. doi:10.1016/j.cell.
2004.09.011.
194. Szentandrássy N, Nagy D, Ruzsnavszky F, Harmati G, Bányász T,
Magyar J, Szentmiklo si AJ, Nánási PP. Powerful technique to test 208. Thiel WH, Chen B, Hund TJ, Koval OM, Purohit A, Song LS, Mohler
selectivity of agents acting on cardiac ion channels: the action PJ, Anderson ME. Proarrhythmic defects in Timothy syndrome
potential voltage-clamp. Curr Med Chem 18: 3737–3756, 2011. require calmodulin kinase II. Circulation 118: 2225–2234, 2008.
doi:10.2174/092986711796642418. doi:10.1161/CIRCULATIONAHA.108.788067.

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1143


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

209. Navedo MF, Cheng EP, Yuan C, Votaw S, Molkentin JD, Scott JD, 224. Spector PS, Curran ME, Zou A, Keating MT, Sanguinetti MC. Fast
Santana LF. Increased coupled gating of L-type Ca21 channels dur- inactivation causes rectification of the IKr channel. J Gen Physiol
ing hypertension and Timothy syndrome. Circ Res 106: 748–756, 107: 611–619, 1996. doi:10.1085/jgp.107.5.611.
2010. doi:10.1161/CIRCRESAHA.109.213363.
225. Yang T, Snyders DJ, Roden DM. Rapid inactivation determines the
210. Napolitano C, Splawski I, Timothy KW, Bloise R, Priori SG. Timothy rectification and [K1]o dependence of the rapid component of the
syndrome. In: GeneReviews, edited by Adam MP, Ardinger HH, delayed rectifier K1 current in cardiac cells. Circ Res 80: 782–789,
Pagon RA, Wallace SE, Bean LJ, Stephens K, Amemiya A. Seattle, 1997. doi:10.1161/01.res.80.6.782.
WA: Univ. of Washington, 1993.
226. Clark RB, Mangoni ME, Lueger A, Couette B, Nargeot J, Giles WR.
211. Zhou Z, Lipsius SL. T-type calcium current in latent pacemaker cells A rapidly activating delayed rectifier K1 current regulates pace-
isolated from cat right atrium. J Mol Cell Cardiol 26: 1211–1219, maker activity in adult mouse sinoatrial node cells. Am J Physiol
1994. doi:10.1006/jmcc.1994.1139. Heart Circ Physiol 286: H1757–H1766, 2004. doi:10.1152/ajpheart.
00753.2003.
212. Bohn G, Moosmang S, Conrad H, Ludwig A, Hofmann F, Klugbauer
N. Expression of T- and L-type calcium channel mRNA in murine 227. Jurkiewicz NK, Sanguinetti MC. Rate-dependent prolongation of
sinoatrial node. FEBS Lett 481: 73–76, 2000. doi:10.1016/S0014- cardiac action potentials by a methanesulfonanilide class III antiar-
5793(00)01979-7. rhythmic agent. Specific block of rapidly activating delayed rectifier
K1 current by dofetilide. Circ Res 72: 75–83, 1993. doi:10.1161/01.
213. Massie BM. Mibefradil, a T-type channel-selective calcium an- res.72.1.75.
tagonist: clinical trials in chronic stable angina pectoris. Am J
Hypertens 11: 95S–102S, 1998. doi:10.1016/s0895-7061(98)  A, Baláti B, Iost N, Takács J, Virág L, Lathrop DA, Csaba L,
228. Varro
00006-5. Tálosi L, Papp JG. The role of the delayed rectifier component IKs
in dog ventricular muscle and Purkinje fibre repolarization. J
214. Sanguinetti MC, Jurkiewicz NK. Two components of cardiac Physiol 523: 67–81, 2000. doi:10.1111/j.1469-7793.2000.00067.x.
delayed rectifier K1 current. Differential sensitivity to block by class
229. Magyar J, Iost N, Ko€ rtve
ly A, Bányász T, Virág L, Szigligeti P, Varro

III antiarrhythmic agents. J Gen Physiol 96: 195–215, 1990.
A, Opincariu M, Sze csi J, Papp JG, Nánási PP. Effects of endothelin-
doi:10.1085/jgp.96.1.195.
1 on calcium and potassium currents in undiseased human ventricu-
215. Nerbonne JM. Molecular basis of functional myocardial potassium lar myocytes. Pflugers Arch 441: 144–149, 2000. doi:10.1007/
channel diversity. Card Electrophysiol Clin 8: 257–273, 2016. s004240000400.
doi:10.1016/j.ccep.2016.01.001.
230. Karle CA, Zitron E, Zhang W, Katho€ fer S, Schoels W, Kiehn J. Rapid
216. Vandenberg JI, Perry MD, Perrin MJ, Mann SA, Ke Y, Hill AP. hERG component IKr of the guinea-pig cardiac delayed rectifier K1 cur-
K1 channels: structure, function, and clinical significance. Physiol rent is inhibited by beta1-adrenoreceptor activation, via cAMP/pro-
Rev 92: 1393–1478, 2012. doi:10.1152/physrev.00036.2011. tein kinase A-dependent pathways. Cardiovasc Res 53: 355–362,
2002. doi:10.1016/s0008-6363(01)00509-0.
217. Nerbonne JM, Kass RS. Molecular physiology of cardiac repolariza-
tion. Physiol Rev 85: 1205–1253, 2005. doi:10.1152/physrev. 231. Wang S, Xu D, Wu TT, Guo Y, Chen YH, Zou JG. Beta1-adrenergic
00002.2005. regulation of rapid component of delayed rectifier K1 currents in
guinea-pig cardiac myocytes. Mol Med Rep 9: 1923–1928, 2014.
218. Abbott GW, Sesti F, Splawski I, Buck ME, Lehmann MH, Timothy doi:10.3892/mmr.2014.2035.
KW, Keating MT, Goldstein SA. MiRP1 forms IKr potassium channels
with HERG and is associated with cardiac arrhythmia. Cell 97: 175– 232. Wang S, Xu DJ, Cai JB, Huang YZ, Zou JG, Cao KJ. Rapid compo-
187, 1999. doi:10.1016/s0092-8674(00)80728-x. nent IKr of cardiac delayed rectifier potassium currents in guinea-
pig is inhibited by alpha1-adrenoreceptor activation via protein ki-
219. Jones DK, Liu F, Vaidyanathan R, Eckhardt LL, Trudeau MC, nase A and protein kinase C-dependent pathways. Eur J
Robertson GA. hERG 1b is critical for human cardiac repolarization. Pharmacol 608: 1–6, 2009. doi:10.1016/j.ejphar.2009.02.017.
Proc Natl Acad Sci USA 111: 18073–18077, 2014. doi:10.1073/
 G. Effects of low [K1]o on the electrical activity of human
233. Christe
pnas.1414945111.
cardiac ventricular and Purkinje cells. Cardiovasc Res 17: 243–250,
220. Trudeau MC, Leung LM, Roti ER, Robertson GA. hERG1a N-terminal 1983. doi:10.1093/cvr/17.4.243.
eag domain-containing polypeptides regulate homomeric hERG1b
234. Firek L, Giles WR. Outward currents underlying repolarization in
and heteromeric hERG1a/hERG1b channels: a possible mechanism
human atrial myocytes. Cardiovasc Res 30: 31–38, 1995.
for long QT syndrome. J Gen Physiol 138: 581–592, 2011. doi:10.
1085/jgp.201110683. 235. Sanguinetti MC, Jurkiewicz NK. Role of external Ca21 and K1 in
gating of cardiac delayed rectifier K1 currents. Pflugers Arch 420:
221. Jost N, Vira´g L, Bitay M, Taka´cs J, Lengyel C, Biliczki P, Nagy Z,
180–186, 1992. doi:10.1007/BF00374988.
Boga´ts G, Lathrop DA, Papp JG, Varro´ A. Restricting excessive
cardiac action potential and QT prolongation: a vital role for IKs in 236. Yang T, Roden DM. Extracellular potassium modulation of drug
human ventricular muscle. Circulation 112: 1392–1399, 2005. doi: block of IKr. Implications for torsade de pointes and reverse use-
10.1161/CIRCULATIONAHA.105.550111. dependence. Circulation 93: 407–411, 1996. doi:10.1161/01.cir.93.
3.407.
csi J, Varro
222. Iost N, Virág L, Opincariu M, Sze  A, Papp JG. Delayed
rectifier potassium current in undiseased human ventricular myo- 237. Be rube
 J, Chahine M, Daleau P. Modulation of HERG potassium
cytes. Cardiovasc Res 40: 508–515, 1998. doi:10.1016/s0008-6363 channel properties by external pH. Pflugers Arch 438: 419–422,
(98)00204-1. 1999. doi:10.1007/s004240050930.

223. Smith PL, Baukrowitz T, Yellen G. The inward rectification mecha- 238. Vereecke J, Carmeliet E. The effect of external pH on the delayed
nism of the HERG cardiac potassium channel. Nature 379: 833– rectifying K1 current in cardiac ventricular myocytes. Pflugers Arch
836, 1996. doi:10.1038/379833a0. 439: 739–751, 2000. doi:10.1007/s004249900243.

1144 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

239. Du CY, El Harchi A, McPate MJ, Orchard CH, Hancox JC. Enhanced 253. Virág L, Iost N, Opincariu M, Szolnoky J, Sze  csi J, Bogáts G,
inhibitory effect of acidosis on hERG potassium channels that incor- Szenohradszky P, Varro  A, Papp JG. The slow component of the
porate the hERG1b isoform. Biochem Biophys Res Commun 405: delayed rectifier potassium current in undiseased human ventricu-
222–227, 2011. doi:10.1016/j.bbrc.2011.01.014. lar myocytes. Cardiovasc Res 49: 790–797, 2001. doi:10.1016/
s0008-6363(00)00306-0.
240. Jiang M, Cabo C, Yao J, Boyden PA, Tseng G. Delayed rectifier K
currents have reduced amplitudes and altered kinetics in myocytes 254. Jost N, Papp JG, Varro  A. Slow delayed rectifier potassium current
from infarcted canine ventricle. Cardiovasc Res 48: 34–43, 2000. (IKs) and the repolarization reserve. Ann Noninvasive Electrocardiol
doi:10.1016/s0008-6363(00)00159-0. 12: 64–78, 2007. doi:10.1111/j.1542-474X.2007.00140.x.

241. Janse MJ. Electrophysiological changes in heart failure and their 255. Volders PG, Stengl M, van Opstal JM, Gerlach U, Spa €tjens RL,
relationship to arrhythmogenesis. Cardiovasc Res 61: 208–217, Beekman JD, Sipido KR, Vos MA. Probing the contribution of IKs to
2004. doi:10.1016/j.cardiores.2003.11.018. canine ventricular repolarization: key role for beta-adrenergic re-
ceptor stimulation. Circulation 107: 2753–2760, 2003. doi:10.1161/
242. Froese A, Breher SS, Waldeyer C, Schindler RF, Nikolaev VO, 01.CIR.0000068344.54010.B3.
Rinne S, Wischmeyer E, Schlueter J, Becher J, Simrick S, Vauti F,
Kuhtz J, Meister P, Kreissl S, Torlopp A, Liebig SK, Laakmann S, 256. Han W, Wang Z, Nattel S. Slow delayed rectifier current and repola-
rization in canine cardiac Purkinje cells. Am J Physiol Heart Circ
M€uller TD, Neumann J, Stieber J, Ludwig A, Maier SK, Decher N,
Physiol 280: H1075–H1080, 2001. doi:10.1152/ajpheart.2001.280.3.
Arnold HH, Kirchhof P, Fabritz L, Brand T. Popeye domain contain-
H1075.
ing proteins are essential for stress-mediated modulation of cardiac
pacemaking in mice. J Clin Invest 122: 1119–1130, 2012. doi:10.1172/ 257. Roden DM. Long QT syndrome: reduced repolarization reserve
JCI59410. and the genetic link. J Intern Med 259: 59–69, 2006. doi:10.1111/
j.1365-2796.2005.01589.x.
243. Richard P, Charron P, Carrier L, Ledeuil C, Cheav T, Pichereau C,
Benaiche A, Isnard R, Dubourg O, Burban M, Gueffet JP, Millaire A, 258. Roden DM. Taking the “idio” out of “idiosyncratic”: predicting tor-
Desnos M, Schwartz K, Hainque B, Komajda M; EUROGENE Heart sades de pointes. Pacing Clin Electrophysiol 21: 1029–1034, 1998.
Failure Project. Hypertrophic cardiomyopathy: distribution of dis- doi:10.1111/j.1540-8159.1998.tb00148.x.
ease genes, spectrum of mutations, and implications for a molecu-
lar diagnosis strategy. Circulation 107: 2227–2232, 2003. doi:10. 259. Varkevisser R, Wijers SC, van der Heyden MA, Beekman JD, Meine
1161/01.CIR.0000066323.15244.54. M, Vos MA. Beat-to-beat variability of repolarization as a new bio-
marker for proarrhythmia in vivo. Heart Rhythm 9: 1718–1726, 2012.
244. Campuzano O, Sarquella-Brugada G, Cesar S, Arbelo E, Brugada J, doi:10.1016/j.hrthm.2012.05.016.
Brugada R. Recent advances in short QT syndrome. Front
 I. Cardiac ventricular repolarization reserve: a prin-
260. Varro A, Baczko
Cardiovasc Med 5: 149, 2018. doi:10.3389/fcvm.2018.00149.
ciple for understanding drug-related proarrhythmic risk. Br J
245. Khera S, Jacobson JT. Short QT syndrome in current clinical prac- Pharmacol 164: 14–36, 2011. doi:10.1111/j.1476-5381.2011.01367.x.
tice. Cardiol Rev 24: 190–193, 2016. doi:10.1097/CRD.0000000
261. Bosch RF, Schneck AC, Csillag S, Eigenberger B, Gerlach U,
000000091.
Brendel J, Lang HJ, Mewis C, Go€gelein H, Seipel L, K€
uhlkamp V.
246. Schwartz PJ, Ackerman MJ, Antzelevitch C, Bezzina CR, Borggrefe Effects of the chromanol HMR 1556 on potassium currents in atrial
M, Cuneo BF, Wilde AA. Inherited cardiac arrhythmias. Nat Rev Dis myocytes. Naunyn Schmiedebergs Arch Pharmacol 367: 281–
Primers 6: 58, 2020. doi:10.1038/s41572-020-0188-7. 288, 2003. doi:10.1007/s00210-002-0672-5.

247. Barhanin J, Lesage F, Guillemare E, Fink M, Lazdunski M, Romey G. 262. Zankov DP, Salloum FN, Jiang M, Tseng GN. Chronic in vivo angio-
KVLQT1 and lsK (minK) proteins associate to form the IKs cardiac po- tensin II administration differentially modulates the slow delayed
rectifier channels in atrial and ventricular myocytes. Heart Rhythm
tassium current. Nature 384: 78–80, 1996. doi:10.1038/384078a0.
16: 108–116, 2019. doi:10.1016/j.hrthm.2018.07.036.
248. Sanguinetti MC, Curran ME, Zou A, Shen J, Spector PS, Atkinson
263. F€ € p L, Bányász T, Szabo
ulo  G, To th IB, Bíro
 T, Lôrincz I, Balogh A,
DL, Keating MT. Coassembly of KVLQT1 and minK (IsK) proteins to
Petô K, Miko I, Nánási PP. Effects of sex hormones on ECG parame-
form cardiac IKs potassium channel. Nature 384: 80–83, 1996.
ters and expression of cardiac ion channels in dogs. Acta Physiol
doi:10.1038/384080a0.
(Oxf) 188: 163–171, 2006. doi:10.1111/j.1748-1716.2006.01618.x.
249. Go€ gelein H, Br€uggemann A, Gerlach U, Brendel J, Busch AE.
264. Li Y, Chen L, Kass RS, Dessauer CW. The A-kinase anchoring pro-
Inhibition of IKs channels by HMR 1556. Naunyn Schmiedebergs tein Yotiao facilitates complex formation between adenylyl cyclase
Arch Pharmacol 362: 480–488, 2000. doi:10.1007/s002100 type 9 and the IKs potassium channel in heart. J Biol Chem 287:
000284. 29815–29824, 2012. doi:10.1074/jbc.M112.380568.
250. Lengyel C, Varro  A, Tábori K, Papp JG, Baczko
 I. Combined phar- 265. Odening KE, Koren G. How do sex hormones modify arrhythmo-
macological block of IKr and IKs increases short-term QT interval genesis in long QT syndrome? Sex hormone effects on arrhythmo-
variability and provokes torsades de pointes. Br J Pharmacol 151: genic substrate and triggered activity. Heart Rhythm 11: 2107–2115,
941–951, 2007. doi:10.1038/sj.bjp.0707297. 2014. doi:10.1016/j.hrthm.2014.06.023.
251. Salata JJ, Jurkiewicz NK, Jow B, Folander K, Guinosso PJ Jr, 266. Lo CF, Numann R. Independent and exclusive modulation of car-
Raynor B, Swanson R, Fermini B. IK of rabbit ventricle is composed diac delayed rectifying K1 current by protein kinase C and protein
of two currents: evidence for IKs. Am J Physiol Heart Circ Physiol kinase A. Circ Res 83: 995–1002, 1998. doi:10.1161/01.res.83.10.
271: H2477–H2489, 1996. doi:10.1152/ajpheart.1996.271.6.H2477. 995.

252. Li GR, Feng J, Yue L, Carrier M, Nattel S. Evidence for two compo- 267. Toda H, Ding WG, Yasuda Y, Toyoda F, Ito M, Matsuura H, Horie M.
nents of delayed rectifier K1 current in human ventricular myo- Stimulatory action of protein kinase C(epsilon) isoform on the slow
cytes. Circ Res 78: 689–696, 1996. doi:10.1161/01.RES.78.4.689. component of delayed rectifier K1 current in guinea-pig atrial

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1145


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

myocytes. Br J Pharmacol 150: 1011–1021, 2007. doi:10.1038/sj.bjp. 280. Moreno C, Oliveras A, de la Cruz A, Bartolucci C, Mun~oz C, Salar E,
0707191. Gimeno JR, Severi S, Comes N, Felipe A, González T, Lambiase P,
Valenzuela C. A new KCNQ1 mutation at the S5 segment that
268. Xiao GQ, Mochly-Rosen D, Boutjdir M. PKC isozyme selective regu- impairs its association with KCNE1 is responsible for short QT syn-
lation of cloned human cardiac delayed slow rectifier K current. drome. Cardiovasc Res 107: 613–623, 2015. doi:10.1093/cvr/
Biochem Biophys Res Commun 306: 1019–1025, 2003. doi:10. cvv196.
1016/s0006-291x(03)01095-7.
281. Zhou X, Bueno-Orovio A, Schilling RJ, Kirkby C, Denning C,
269. Salata JJ, Jurkiewicz NK, Wang J, Evans BE, Orme HT, Sanguinetti Rajamohan D, Burrage K, Tinker A, Rodriguez B, Harmer SC.
MC. A novel benzodiazepine that activates cardiac slow delayed Investigating the complex arrhythmic phenotype caused by the
rectifier K1 currents. Mol Pharmacol 54: 220–230, 1998. doi:10. gain-of-function mutation KCNQ1-G229D. Front Physiol 10: 259,
1124/mol.54.1.220. 2019. doi:10.3389/fphys.2019.00259.

270. Bartos DC, Morotti S, Ginsburg KS, Grandi E, Bers DM. Quantitative 282. Gadsby DC, Cranefield PF. Electrogenic sodium extrusion in car-
analysis of the Ca21-dependent regulation of delayed rectifier K1 diac Purkinje fibers. J Gen Physiol 73: 819–837, 1979. doi:10.1085/
current IKs in rabbit ventricular myocytes. J Physiol 595: 2253– jgp.73.6.819.
2268, 2017. doi:10.1113/JP273676.
283. Hirano Y, Hiraoka M. Changes in K1 currents induced by Ba21 in
 T, Jost N, Magyar J, Biliczki P, Kocsis E,
271. Lengyel C, Virág L, Biro guinea pig ventricular muscles. Am J Physiol Heart Circ Physiol
Skoumal R, Nánási PP, To  th M, Kecskeme ti V, Papp JG, Varro
 A. 251: H24–H33, 1986. doi:10.1152/ajpheart.1986.251.1.H24.
Diabetes mellitus attenuates the repolarization reserve in mamma-
lian heart. Cardiovasc Res 73: 512–520, 2007. doi:10.1016/j. 284. Martin RL, Koumi S, Ten Eick RE. Comparison of the effects of inter-
cardiores.2006.11.010. nal [Mg21] on IK1 in cat and guinea-pig cardiac ventricular myo-
cytes. J Mol Cell Cardiol 27: 673–691, 1995. doi:10.1016/S0022-
272. Stengl M, Ramakers C, Donker DW, Nabar A, Rybin AV, Spa €tjens 2828(08)80059-4.
RL, van der Nagel T, Wodzig WK, Sipido KR, Antoons G, Moorman
285. Matsuda H. Effects of external and internal K1 ions on magnesium
AF, Vos MA, Volders PG. Temporal patterns of electrical remodel-
block of inwardly rectifying K1 channels in guinea-pig heart cells. J
ing in canine ventricular hypertrophy: focus on IKs downregulation
Physiol 435: 83–99, 1991. doi:10.1113/jphysiol.1991.sp018499.
and blunted beta-adrenergic activation. Cardiovasc Res 72: 90–
100, 2006. doi:10.1016/j.cardiores.2006.07.015. 286. Biermans G, Vereecke J, Carmeliet E. The mechanism of the inacti-
vation of the inward-rectifying K current during hyperpolarizing
273. Bartos DC, Anderson JB, Bastiaenen R, Johnson JN, Gollob MH,
steps in guinea-pig ventricular myocytes. Pflugers Arch 410: 604–
Tester DJ, Burgess DE, Homfray T, Behr ER, Ackerman MJ,
613, 1987. doi:10.1007/BF00581320.
Guicheney P, Delisle BP. A KCNQ1 mutation causes a high pene-
trance for familial atrial fibrillation. J Cardiovasc Electrophysiol 24: 287. Nagy N, Acsai K, Kormos A, Sebo†k Z, Farkas AS, Jost N, Nánási PP,
562–569, 2013. doi:10.1111/jce.12068. Papp JG, Varro  A, To th A. [Ca21]i-induced augmentation of the
inward rectifier potassium current (IK1) in canine and human ventric-
274. Bartos DC, Duchatelet S, Burgess DE, Klug D, Denjoy I, Peat R,
ular myocardium. Pflugers Arch 465: 1621–1635, 2013. doi:10.1007/
Lupoglazoff JM, Fressart V, Berthet M, Ackerman MJ, January CT, s00424-013-1309-x.
Guicheney P, Delisle BP. R231C mutation in KCNQ1 causes long QT
syndrome type 1 and familial atrial fibrillation. Heart Rhythm 8: 48– 288. Noble D, Tsien RW. Outward membrane currents activated in the
55, 2011. doi:10.1016/j.hrthm.2010.09.010. plateau range of potentials in cardiac Purkinje fibres. J Physiol
200: 205–231, 1969. doi:10.1113/jphysiol.1969.sp008689.
275. Chen YH, Xu SJ, Bendahhou S, Wang XL, Wang Y, Xu WY, Jin HW,
Sun H, Su XY, Zhuang QN, Yang YQ, Li YB, Liu Y, Xu HJ, Li XF, Ma 289. Fauconnier J, Lacampagne A, Rauzier JM, Vassort G, Richard S.
N, Mou CP, Chen Z, Barhanin J, Huang W. KCNQ1 gain-of-function Ca21-dependent reduction of IK1 in rat ventricular cells: a novel par-
mutation in familial atrial fibrillation. Science 299: 251–254, 2003. adigm for arrhythmia in heart failure? Cardiovasc Res 68: 204–212,
doi:10.1126/science.1077771. 2005. doi:10.1016/j.cardiores.2005.05.024.

276. Das S, Makino S, Melman YF, Shea MA, Goyal SB, Rosenzweig A, 290. Zaza A, Rocchetti M, Brioschi A, Cantadori A, Ferroni A. Dynamic
Macrae CA, Ellinor PT. Mutation in the S3 segment of KCNQ1 Ca21-induced inward rectification of K1 current during the ventricu-
results in familial lone atrial fibrillation. Heart Rhythm 6: 1146–1153, lar action potential. Circ Res 82: 947–956, 1998. doi:10.1161/01.
2009. doi:10.1016/j.hrthm.2009.04.015. res.82.9.947.

277. Hong K, Piper DR, Diaz-Valdecantos A, Brugada J, Oliva A, 291. Dhamoon AS, Jalife J. The inward rectifier current (IK1) controls car-
Burashnikov E, Santos-de-Soto J, Grueso-Montero J, Diaz-Enfante diac excitability and is involved in arrhythmogenesis. Heart
E, Brugada P, Sachse F, Sanguinetti MC, Brugada R. De novo Rhythm 2: 316–324, 2005. doi:10.1016/j.hrthm.2004.11.012.
KCNQ1 mutation responsible for atrial fibrillation and short QT syn-
292. Lopatin AN, Nichols CG. [K1] dependence of polyamine-induced
drome in utero. Cardiovasc Res 68: 433–440, 2005. doi:10.1016/j.
rectification in inward rectifier potassium channels (IRK1, Kir2.1). J
cardiores.2005.06.023.
Gen Physiol 108: 105–113, 1996. doi:10.1085/jgp.108.2.105.
278. Ki CS, Jung CL, Kim HJ, Baek KH, Park SJ, On YK, Kim KS, Noh SJ,
293. Nichols CG, Lopatin AN. Inward rectifier potassium channels. Annu
Youm JB, Kim JS, Cho H. A KCNQ1 mutation causes age-depend-
Rev Physiol 59: 171–191, 1997. doi:10.1146/annurev.physiol.59.1.171.
ant bradycardia and persistent atrial fibrillation. Pflugers Arch 466:
529–540, 2014. doi:10.1007/s00424-013-1337-6. 294. Weiss JN, Qu Z, Shivkumar K. Electrophysiology of hypokalemia
and hyperkalemia. Circ Arrhythm Electrophysiol 10: e004667,
279. Lundby A, Ravn LS, Svendsen JH, Olesen SP, Schmitt N. KCNQ1 2017. doi:10.1161/CIRCEP.116.004667.
mutation Q147R is associated with atrial fibrillation and prolonged
QT interval. Heart Rhythm 4: 1532–1541, 2007. doi:10.1016/j. € fer S, Thomas
295. Karle CA, Zitron E, Zhang W, Wendt-Nordahl G, Katho
hrthm.2007.07.022. D, Gut B, Scholz E, Vahl CF, Katus HA, Kiehn J. Human cardiac

1146 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

inwardly-rectifying K1 channel Kir(2.1b) is inhibited by direct protein 309. Wettwer E, Ha´la O, Christ T, Heubach JF, Dobrev D, Knaut M,
kinase C-dependent regulation in human isolated cardiomyocytes Varro´ A, Ravens U. Role of IKur in controlling action potential
and in an expression system. Circulation 106: 1493–1499, 2002. shape and contractility in the human atrium: influence of chronic
doi:10.1161/01.cir.0000029747.53262.5c. atrial fibrillation. Circulation 110: 2299–2306, 2004. doi:10.1161/01.
CIR.0000145155.60288.71.
296. Koumi S, Wasserstrom JA, Ten Eick RE. Beta-adrenergic and cho-
linergic modulation of inward rectifier K1 channel function and 310. Ford J, Milnes J, Wettwer E, Christ T, Rogers M, Sutton K, Madge D,
phosphorylation in guinea-pig ventricle. J Physiol 486: 661–678, Virag L, Jost N, Horvath Z, Matschke K, Varro A, Ravens U. Human
1995. doi:10.1113/jphysiol.1995.sp020842. electrophysiological and pharmacological properties of XEN-
D0101: a novel atrial-selective Kv1.5/IKur inhibitor. J Cardiovasc
297. Veldkamp MW, Geuzebroek GS, Baartscheer A, Verkerk AO, Pharmacol 61: 408–415, 2013. doi:10.1097/FJC.0b013e318287
Schumacher CA, Suarez GG, Berger WR, Casini S, van Amersfoorth 80eb.
SC, Scholman KT, Driessen AH, Belterman CN, van Ginneken AC,
de Groot JR, de Bakker JM, Remme CA, Boukens BJ, Coronel R. 311. Shunmugam SR, Sugihara C, Freemantle N, Round P, Furniss S,
Neurokinin-3 receptor activation selectively prolongs atrial refracto- Sulke N. A double-blind, randomised, placebo-controlled, cross-
riness by inhibition of a background K1 channel. Nat Commun 9: over study assessing the use of XEN-D0103 in patients with parox-
4357, 2018. doi:10.1038/s41467-018-06530-5. ysmal atrial fibrillation and implanted pacemakers allowing continu-
ous beat-to-beat monitoring of drug efficacy. J Interv Card
298. Takanari H, Nalos L, Stary-Weinzinger A, de Git KC, Varkevisser R, Electrophysiol 51: 191–197, 2018. doi:10.1007/s10840-018-0318-2.
Linder T, Houtman MJ, Peschar M, de Boer TP, Tidwell RR, Rook
MB, Vos MA, van der Heyden MA. Efficient and specific cardiac IK1 312. Eisner DA, Vaughan-Jones RD. Do calcium-activated potassium
inhibition by a new pentamidine analogue. Cardiovasc Res 99: channels exist in the heart? Cell Calcium 4: 371–386, 1983.
203–214, 2013. doi:10.1093/cvr/cvt103. doi:10.1016/0143-4160(83)90015-5.

299. Veerapandiyan A, Statland JM, Tawil , R. Andersen-Tawil syn- 313. Xu Y, Tuteja D, Zhang Z, Xu D, Zhang Y, Rodriguez J, Nie L, Tuxson
drome. In: GeneReviews, edited by Adam MP, Ardinger HH, Pagon HR, Young JN, Glatter KA, Vázquez AE, Yamoah EN,
RA, Wallace SE, Bean LJ, Stephens K, Amemiya A. Seattle, WA: Chiamvimonvat N. Molecular identification and functional roles of a
Univ. of Washington, 1993. Ca21-activated K1 channel in human and mouse hearts. J Biol
Chem 278: 49085–49094, 2003. doi:10.1074/jbc.M307508200.
300. Tsuboi M, Antzelevitch C. Cellular basis for electrocardiographic
and arrhythmic manifestations of Andersen-Tawil syndrome (LQT7). 314. Skibsbye L, Poulet C, Diness JG, Bentzen BH, Yuan L, Kappert U,
Heart Rhythm 3: 328–335, 2006. doi:10.1016/j.hrthm.2005.11.026. Matschke K, Wettwer E, Ravens U, Grunnet M, Christ T, Jespersen
T. Small-conductance calcium-activated potassium (SK) channels
301. Tamkun MM, Knoth KM, Walbridge JA, Kroemer H, Roden DM, contribute to action potential repolarization in human atria.
Glover DM. Molecular cloning and characterization of two voltage- Cardiovasc Res 103: 156–167, 2014. doi:10.1093/cvr/cvu121.
gated K1 channel cDNAs from human ventricle. FASEB J 5: 331–
337, 1991. doi:10.1096/fasebj.5.3.2001794. 315. Chang PC, Turker I, Lopshire JC, Masroor S, Nguyen BL, Tao W,
Rubart M, Chen PS, Chen Z, Ai T. Heterogeneous upregulation of
302. Fedida D, Wible B, Wang Z, Fermini B, Faust F, Nattel S, Brown AM. apamin-sensitive potassium currents in failing human ventricles. J
Identity of a novel delayed rectifier current from human heart with a Am Heart Assoc 2: e004713, 2013. doi:10.1161/JAHA.112.004713.
cloned K1 channel current. Circ Res 73: 210–216, 1993. doi:10.1161/
01.res.73.1.210. 316. Kirchhoff JE, Diness JG, Sheykhzade M, Grunnet M, Jespersen T.
Synergistic antiarrhythmic effect of combining inhibition of Ca21-
303. Amos GJ, Wettwer E, Metzger F, Li Q, Himmel HM, Ravens U. activated K1 (SK) channels and voltage-gated Na1 channels in an
Differences between outward currents of human atrial and subepi- isolated heart model of atrial fibrillation. Heart Rhythm 12: 409–
cardial ventricular myocytes. J Physiol 491: 31–50, 1996. doi:10.1113/ 418, 2015. doi:10.1016/j.hrthm.2014.12.010.
jphysiol.1996.sp021194.
317. Nattel S. Calcium-activated potassium current: a novel ion channel
304. Majumder K, De Biasi M, Wang Z, Wible BA. Molecular cloning and candidate in atrial fibrillation. J Physiol 587: 1385–1386, 2009.
functional expression of a novel potassium channel beta-subunit doi:10.1113/jphysiol.2009.170621.
from human atrium. FEBS Lett 361: 13–16, 1995. doi:10.1016/0014-
5793(95)00120-x. 318. Hamilton S, Polina I, Terentyeva R, Bronk P, Kim TY, Roder K,
Clements RT, Koren G, Choi BR, Terentyev D. PKA phosphorylation
305. Nattel S. The molecular and ionic specificity of antiarrhythmic drug underlies functional recruitment of sarcolemmal SK2 channels in
actions. J Cardiovasc Electrophysiol 10: 272–282, 1999. doi:10.1111/ ventricular myocytes from hypertrophic hearts. J Physiol 598:
j.1540-8167.1999.tb00673.x. 2847–2873, 2020. doi:10.1113/JP277618.

306. Yue L, Feng J, Li GR, Nattel S. Characterization of an ultrarapid 319. Fan X, Yu Y, Lan H, Ou X, Yang L, Li T, Cao J, Zeng X, Li M. Ca21/
delayed rectifier potassium channel involved in canine atrial repola- calmodulin-dependent protein kinase II (CaMKII) increases small-
rization. J Physiol 496: 647–662, 1996. doi:10.1113/jphysiol.1996. conductance Ca21-activated K1 current in patients with chronic
sp021716. atrial fibrillation. Med Sci Monit 24: 3011–3023, 2018. doi:10.12659/
MSM.909684.
307. Yue L, Feng J, Wang Z, Nattel S. Adrenergic control of the ultra-
rapid delayed rectifier current in canine atrial myocytes. J Physiol 320. Xia XM, Fakler B, Rivard A, Wayman G, Johnson-Pais T, Keen JE,
516: 385–398, 1999. doi:10.1111/j.1469-7793.1999.0385v.x. Ishii T, Hirschberg B, Bond CT, Lutsenko S, Maylie J, Adelman JP.
Mechanism of calcium gating in small-conductance calcium-acti-
308. Sridhar A, da Cunha DN, Lacombe VA, Zhou Q, Fox JJ, Hamlin RL, vated potassium channels. Nature 395: 503–507, 1998. doi:
Carnes CA. The plateau outward current in canine ventricle, sensi- 10.1038/26758.
tive to 4-aminopyridine, is a constitutive contributor to ventricular
repolarization. Br J Pharmacol 152: 870–879, 2007. doi:10.1038/sj. 321. Nagy N, Szuts V, Horváth Z, Sepre nyi G, Farkas AS, Acsai K, Prorok
bjp.0707403.  A, To
J, Bitay M, Kun A, Pataricza J, Papp JG, Nánási PP, Varro  th A.

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1147


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

Does small-conductance calcium-activated potassium channel con- 336. Chi LG, Tamura Y, Hoff PT, Macha M, Gallagher KP, Schork MA,
tribute to cardiac repolarization? J Mol Cell Cardiol 47: 656–663, Lucchesi BR. Effect of superoxide dismutase on myocardial infarct
2009. doi:10.1016/j.yjmcc.2009.07.019. size in the canine heart after 6 hours of regional ischemia and
reperfusion: a demonstration of myocardial salvage. Circ Res 64:
322. Nagy N, Márton Z, Kiss L, Varro A, Nánási PP, To  th A. Role of Ca21- 665–675, 1989. doi:10.1161/01.res.64.4.665.
sensitive K1 currents in controlling ventricular repolarization: possi-
ble implications for future antiarrhythmic drug therapy. Curr Med 337. del Valle HF, Lascano EC, Negroni JA, Crottogini AJ. Glibenclamide
Chem 18: 3622–3639, 2011. doi:10.2174/092986711796642463. effects on reperfusion-induced malignant arrhythmias and left ven-
tricular mechanical recovery from stunning in conscious sheep.
323. Reher TA, Wang Z, Hsueh CH, Chang PC, Pan Z, Kumar M, Patel J, Cardiovasc Res 50: 474–485, 2001. doi:10.1016/S0008-6363(01)
Tan J, Shen C, Chen Z, Fishbein MC, Rubart M, Boyden P, Chen PS. 00209-7.
Small-conductance calcium-activated potassium current in normal
rabbit cardiac Purkinje cells. J Am Heart Assoc 6: e005471, 2017. 338. Carlsson L, Abrahamsson C, Drews L, Duker G. Antiarrhythmic
doi:10.1161/JAHA.117.005471. effects of potassium channel openers in rhythm abnormalities
related to delayed repolarization. Circulation 85: 1491–1500, 1992.
324. Duan DY, Fermini B, Nattel S. Sustained outward current observed doi:10.1161/01.cir.85.4.1491.
after Ito1 inactivation in rabbit atrial myocytes is a novel Cl current.
Am J Physiol Heart Circ Physiol 263: H1967–H1971, 1992. 339. Spinelli W, Sorota S, Siegal M, Hoffman BF. Antiarrhythmic actions
doi:10.1152/ajpheart.1992.263.6.H1967. of the ATP-regulated K1 current activated by pinacidil. Circ Res 68:
1127–1137, 1991. doi:10.1161/01.res.68.4.1127.
325. Noma A. ATP-regulated K1 channels in cardiac muscle. Nature
305: 147–148, 1983. doi:10.1038/305147a0. 340. Wolk R, Cobbe SM, Kane KA, Hicks MN. Relevance of inter- and in-
traventricular electrical dispersion to arrhythmogenesis in normal
326. Inagaki N, Gonoi T, Clement JP, Namba N, Inazawa J, Gonzalez G, and ischaemic rabbit myocardium: a study with cromakalim, 5-
Aguilar-Bryan L, Seino S, Bryan J. Reconstitution of IKATP: an hydroxydecanoate and glibenclamide. J Cardiovasc Pharmacol
inward rectifier subunit plus the sulfonylurea receptor. Science 33: 323–334, 1999. doi:10.1097/00005344-199902000-00022.
270: 1166–1170, 1995. doi:10.1126/science.270.5239.1166.
341. Bril A, Man RY. Effects of the potassium channel activator, BRL
327. Chutkow WA, Simon MC, Le Beau MM, Burant CF. Cloning, tissue 34915, on the action potential characteristics of canine cardiac
expression, and chromosomal localization of SUR2, the putative Purkinje fibers. J Pharmacol Exp Ther 253: 1090–1096, 1990.
drug-binding subunit of cardiac, skeletal muscle, and vascular
KATP channels. Diabetes 45: 1439–1445, 1996. doi:10.2337/ 342. Lathrop DA, Nánási PP, Varro  A. In vitro cardiac models of dog
diab.45.10.1439. Purkinje fibre triggered and spontaneous electrical activity: effects
of nicorandil. Br J Pharmacol 99: 119–123, 1990. doi:10.1111/j.1476-
328. Deutsch N, Klitzner TS, Lamp ST, Weiss JN. Activation of cardiac 5381.1990.tb14664.x.
ATP-sensitive K1 current during hypoxia: correlation with tissue
ATP levels. Am J Physiol Heart Circ Physiol 261: H671–H676, 1991. 343. Cole WC, McPherson CD, Sontag D. ATP-regulated K1 channels
doi:10.1152/ajpheart.1991.261.3.H671. protect the myocardium against ischemia/reperfusion damage.
Circ Res 69: 571–581, 1991. doi:10.1161/01.res.69.3.571.
329. Loussouarn G, Marton LJ, Nichols CG. Molecular basis of inward
rectification: structural features of the blocker defined by extended 344. Maddaford TG, Dibrov E, Hurtado C, Pierce GN. Reduced expres-
polyamine analogs. Mol Pharmacol 68: 298–304, 2005. doi:10. sion of the Na1/Ca21 exchanger in adult cardiomyocytes via
1124/mol.105.012377. adenovirally delivered shRNA results in resistance to simulated is-
chemic injury. Am J Physiol Heart Circ Physiol 298: H360–H366,
330. Nichols CG. KATP channels as molecular sensors of cellular metab- 2010. doi:10.1152/ajpheart.00932.2009.
olism. Nature 440: 470–476, 2006. doi:10.1038/nature04711.
345. McPherson CD, Pierce GN, Cole WC. Ischemic cardioprotection by
331. Shyng S, Ferrigni T, Nichols CG. Control of rectification and gating ATP-sensitive K1 channels involves high-energy phosphate preser-
of cloned KATP channels by the Kir6.2 subunit. J Gen Physiol 110: vation. Am J Physiol Heart Circ Physiol 265: H1809–H1818, 1993.
141–153, 1997. doi:10.1085/jgp.110.2.141. doi:10.1152/ajpheart.1993.265.5.H1809.

332. Chi L, Uprichard AC, Lucchesi BR. Profibrillatory actions of pinacidil 346. Baczko  I, Giles WR, Light PE. Pharmacological activation of plasma-
in a conscious canine model of sudden coronary death. J membrane KATP channels reduces reoxygenation-induced Ca21
Cardiovasc Pharmacol 15: 452–464, 1990. doi:10.1097/00005344- overload in cardiac myocytes via modulation of the diastolic mem-
199003000-00016. brane potential. Br J Pharmacol 141: 1059–1067, 2004. doi:10.1038/
sj.bjp.0705702.
333. Billman GE, Englert HC, Scho € lkens BA. HMR 1883, a novel cardiose-
lective inhibitor of the ATP-sensitive potassium channel. Part II: 347. Baczko I, Giles WR, Light PE. Resting membrane potential regulates
effects on susceptibility to ventricular fibrillation induced by myo- Na1-Ca21 exchange-mediated Ca21 overload during hypoxia-
cardial ischemia in conscious dogs. J Pharmacol Exp Ther 286: reoxygenation in rat ventricular myocytes. J Physiol 550: 889–
1465–1473, 1998. 898, 2003. doi:10.1113/jphysiol.2003.043372.

334. Englert HC, Heitsch H, Gerlach U, Knieps S. Blockers of the ATP- 348. Inoue I, Nagase H, Kishi K, Higuti T. ATP-sensitive K1 channel in
sensitive potassium channel SUR2A/Kir6.2: a new approach to pre- the mitochondrial inner membrane. Nature 352: 244–247, 1991.
vent sudden cardiac death. Curr Med Chem Cardiovasc Hematol doi:10.1038/352244a0.
Agents 1: 253–271, 2003. doi:10.2174/1568016033477423.
349. Paucek P, Mironova G, Mahdi F, Beavis AD, Woldegiorgis G, Garlid
335. Vajda S, Baczko I, Leprán I. Selective cardiac plasma-membrane KD. Reconstitution and partial purification of the glibenclamide-sen-
KATP channel inhibition is defibrillatory and improves survival during sitive, ATP-dependent K1 channel from rat liver and beef heart mi-
acute myocardial ischemia and reperfusion. Eur J Pharmacol 577: tochondria. J Biol Chem 267: 26062–26069, 1992. doi:10.1016/
115–123, 2007. doi:10.1016/j.ejphar.2007.08.016. S0021-9258(18)35717-X.

1148 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

350. Light PE, Kanji HD, Fox JE, French RJ. Distinct myoprotective roles 364. Dascal N, Schreibmayer W, Lim NF, Wang W, Chavkin C, DiMagno
of cardiac sarcolemmal and mitochondrial KATP channels during L, Labarca C, Kieffer BL, Gaveriaux-Ruff C, Trollinger D. Atrial G pro-
metabolic inhibition and recovery. FASEB J 15: 2586–2594, 2001. tein-activated K1 channel: expression cloning and molecular prop-
doi:10.1096/fj.01-0188com. erties. Proc Natl Acad Sci USA 90: 10235–10239, 1993. doi:10.
1073/pnas.90.21.10235.
351. Nakayama K, Suzuki Y, Hashimoto K. Sustained atrial fibrillation by
acetylcholine infusion into the sinus node artery. Tohoku J Exp 365. Holmes AP, Yu TY, Tull S, Syeda F, Kuhlmann SM, O’Brien SM,
Med 96: 333–339, 1968. doi:10.1620/tjem.96.333. Patel P, Brain KL, Pavlovic D, Brown NA, Fabritz L, Kirchhof P. A re-
gional reduction in Ito and IKACh in the murine posterior left atrial
352. Corey S, Clapham DE. Identification of native atrial G-protein-regu- myocardium is associated with action potential prolongation and
lated inwardly rectifying K1 (GIRK4) channel homomultimers. J Biol increased ectopic activity. PLoS One 11: e0154077, 2016. doi:
Chem 273: 27499–27504, 1998. doi:10.1074/jbc.273.42.27499. 10.1371/journal.pone.0154077.
353. Nobles M, Montaigne D, Sebastian S, Birnbaumer L, Tinker A. 366. Kuß J, Stallmeyer B, Goldstein M, Rinne S, Pees C, Zumhagen S,
Differential effects of inhibitory G protein isoforms on G protein- Seebohm G, Decher N, Pott L, Kienitz MC, Schulze-Bahr E. Familial
gated inwardly rectifying K1 currents in adult murine atria. Am J sinus node disease caused by a gain of GIRK (G-protein activated
Physiol Cell Physiol 314: C616–C626, 2018. doi:10.1152/ajpcell. inwardly rectifying K1 channel) channel function. Circ Genom
00271.2016. Precis Med 12: e002238, 2019. doi:10.1161/CIRCGEN.118.002238.

354. Kurachi Y. G protein regulation of cardiac muscarinic potassium 367. Hume JR, Harvey RD. Chloride conductance pathways in heart. Am
channel. Am J Physiol Cell Physiol 269: C821–C830, 1995. J Physiol Cell Physiol 261: C399–C412, 1991. doi:10.1152/ajpcell.
doi:10.1152/ajpcell.1995.269.4.C821. 1991.261.3.C399.

355. Moreno-Galindo EG, Alamilla J, Sanchez-Chapula JA, Tristani- 368. Hiraoka M, Kawano S, Hirano Y, Furukawa T. Role of cardiac chlo-
Firouzi M, Navarro-Polanco RA. The agonist-specific voltage de- ride currents in changes in action potential characteristics and
pendence of M2 muscarinic receptors modulates the deactivation arrhythmias. Cardiovasc Res 40: 23–33, 1998. doi:10.1016/s0008-
of the acetylcholine-gated K1 current (I KACh). Pflugers Arch 468: 6363(98)00173-4.
1207–1214, 2016. doi:10.1007/s00424-016-1812-y.
369. Duan D. Phenomics of cardiac chloride channels: the systematic
356. Navarro-Polanco RA, Are  chiga-Figueroa IA, Salazar-Fajardo PD, study of chloride channel function in the heart. J Physiol 587:
Benavides-Haro DE, Rodríguez-Elías JC, Sachse FB, Tristani-Firouzi 2163–2177, 2009. doi:10.1113/jphysiol.2008.165860.
M, Sánchez-Chapula JA, Moreno-Galindo EG. Voltage sensitivity of
M2 muscarinic receptors underlies the delayed rectifier-like activa- 370. Duan DD. Phenomics of cardiac chloride channels. Compr Physiol
tion of ACh-gated K1 current by choline in feline atrial myocytes. J 3: 667–692, 2013. doi:10.1002/cphy.c110014.
Physiol 591: 4273–4286, 2013. doi:10.1113/jphysiol.2013.255166. 371. Baumgarten CM, Clemo HF. Swelling-activated chloride channels
357. Kaseda S, Zipes DP. Supersensitivity to acetylcholine of canine in cardiac physiology and pathophysiology. Prog Biophys Mol Biol
sinus and AV nodes after parasympathetic denervation. Am J 82: 25–42, 2003. doi:10.1016/s0079-6107(03)00003-8.
Physiol Heart Circ Physiol 255: H534–H539, 1988. doi:10.1152/ 372. Dudel J, Peper K, R€
udel R, Trautwein W. The dynamic chloride
ajpheart.1988.255.3.H534. component of membrane current in Purkinje fibers. Pflugers Arch
Gesamte Physiol Menschen Tiere 295: 197–212, 1967. doi:10.1007/
358. Dobrev D, Friedrich A, Voigt N, Jost N, Wettwer E, Christ T, Knaut
BF01844100.
M, Ravens U. The G protein-gated potassium current IK,ACh is con-
stitutively active in patients with chronic atrial fibrillation. 373. Kenyon JL, Gibbons WR. Influence of chloride, potassium, and tet-
Circulation 112: 3697–3706, 2005. doi:10.1161/CIRCULATIONAHA. raethylammonium on the early outward current of sheep cardiac
105.575332. Purkinje fibers. J Gen Physiol 73: 117–138, 1979. doi:10.1085/
jgp.73.2.117.
359. Ravens U. Atrial-selective K1 channel blockers: potential antiar-
rhythmic drugs in atrial fibrillation? Can J Physiol Pharmacol 95: 374. Tseng GN, Hoffman BF. Two components of transient outward cur-
1313–1318, 2017. doi:10.1139/cjpp-2017-0024. rent in canine ventricular myocytes. Circ Res 64: 633–647, 1989.
doi:10.1161/01.res.64.4.633.
360. Bukiya AN, Rosenhouse-Dantsker A. Synergistic activation of G
protein-gated inwardly rectifying potassium channels by choles- 375. Zygmunt AC, Gibbons WR. Properties of the calcium-activated
terol and PI(4,5)P2. Biochim Biophys Acta Biomembr 1859: 1233– chloride current in heart. J Gen Physiol 99: 391–414, 1992.
1241, 2017. doi:10.1016/j.bbamem.2017.03.023. doi:10.1085/jgp.99.3.391.

361. Pfaffinger PJ, Martin JM, Hunter DD, Nathanson NM, Hille B. GTP- 376. F€ € p L, Fiák E, Szentandrássy N, Magyar J, Nánási PP, Bányász T.
ulo
binding proteins couple cardiac muscarinic receptors to a K chan- The role of transmembrane chloride current in afterdepolarisations
nel. Nature 317: 536–538, 1985. doi:10.1038/317536a0. in canine ventricular cardiomyocytes. Gen Physiol Biophys 22:
341–353, 2003.
362. Voigt N, Abu-Taha I, Heijman J, Dobrev D. Constitutive activity of
the acetylcholine-activated potassium current IK,ACh in cardiomyo- 377. Horváth B, Váczi K, Hegyi B, Go € nczi M, Dienes B, Kistámas K,
cytes. Adv Pharmacol 70: 393–409, 2014. doi:10.1016/B978-0-12- Bányász T, Magyar J, Baczko I, Varro  A, Seprenyi G, Csernoch L,
417197-8.00013-4. Nánási PP, Szentandrássy N. Sarcolemmal Ca21-entry through L-
type Ca21 channels controls the profile of Ca21-activated Cl cur-
363. Morishima M, Iwata E, Nakada C, Tsukamoto Y, Takanari H, rent in canine ventricular myocytes. J Mol Cell Cardiol 97: 125–139,
Miyamoto S, Moriyama M, Ono K. Atrial fibrillation-mediated upreg- 2016. doi:10.1016/j.yjmcc.2016.05.006.
ulation of miR-30d regulates myocardial electrical remodeling of
the G-protein-gated K1 channel, IK.ACh. Circ J 80: 1346–1355, 378. O’Driscoll KE, Hatton WJ, Burkin HR, Leblanc N, Britton FC.
2016. doi:10.1253/circj.CJ-15-1276. Expression, localization, and functional properties of Bestrophin 3

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1149


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

channel isolated from mouse heart. Am J Physiol Cell Physiol 295: 394. Duan D, Ye L, Britton F, Horowitz B, Hume JR. UltraRapid communi-
C1610–C1624, 2008. doi:10.1152/ajpcell.00461.2008. cations: a novel anionic inward rectifier in native cardiac myocytes.
Circ Res 86: 485, 2000.
379. O’Driscoll KE, Leblanc N, Hatton WJ, Britton FC. Functional proper-
ties of murine bestrophin 1 channel. Biochem Biophys Res 395. Furukawa T, Ogura T, Katayama Y, Hiraoka M. Characteristics of
Commun 384: 476–481, 2009. doi:10.1016/j.bbrc.2009.05.008. rabbit ClC-2 current expressed in Xenopus oocytes and its contribu-
tion to volume regulation. Am J Physiol Cell Physiol 274: C500–
380. Sun H, Tsunenari T, Yau KW, Nathans J. The vitelliform macular C512, 1998. doi:10.1152/ajpcell.1998.274.2.C500.
dystrophy protein defines a new family of chloride channels. Proc
Natl Acad Sci USA 99: 4008–4013, 2002. doi:10.1073/pnas. 396. Huang ZM, Prasad C, Britton FC, Ye LL, Hatton WJ, Duan D.
052692999. Functional role of CLC-2 chloride inward rectifier channels in car-
diac sinoatrial nodal pacemaker cells. J Mol Cell Cardiol 47: 121–
381. Caputo A, Caci E, Ferrera L, Pedemonte N, Barsanti C, Sondo E, 132, 2009. doi:10.1016/j.yjmcc.2009.04.008.
Pfeffer U, Ravazzolo R, Zegarra-Moran O, Galietta LJ. TMEM16A, a
membrane protein associated with calcium-dependent chloride 397. Yamamoto S, Ehara T. Acidic extracellular pH-activated outwardly
channel activity. Science 322: 590–594, 2008. doi:10.1126/ rectifying chloride current in mammalian cardiac myocytes. Am J
science.1163518. Physiol Heart Circ Physiol 290: H1905–H1914, 2006. doi:10.1152/
ajpheart.00965.2005.
382. Schroeder BC, Cheng T, Jan YN, Jan LY. Expression cloning of
TMEM16A as a calcium-activated chloride channel subunit. Cell 398. Sadoshima J, Izumo S. The cellular and molecular response of car-
134: 1019–1029, 2008. doi:10.1016/j.cell.2008.09.003. diac myocytes to mechanical stress. Annu Rev Physiol 59: 551–
571, 1997. doi:10.1146/annurev.physiol.59.1.551.
383. Yang YD, Cho H, Koo JY, Tak MH, Cho Y, Shim WS, Park SP, Lee J,
Lee B, Kim BM, Raouf R, Shin YK, Oh U. TMEM16A confers recep- 399. Sadoshima J, Qiu Z, Morgan JP, Izumo S. Tyrosine kinase activation
tor-activated calcium-dependent chloride conductance. Nature is an immediate and essential step in hypotonic cell swelling-
455: 1210–1215, 2008. doi:10.1038/nature07313. induced ERK activation and c-fos gene expression in cardiac myo-
cytes. EMBO J 15: 5535–5546, 1996. doi:10.1002/j.1460-2075.
384. Zygmunt AC. Intracellular calcium activates a chloride current in ca- 1996.tb00938.x.
nine ventricular myocytes. Am J Physiol Heart Circ Physiol 267:
H1984–H1995, 1994. doi:10.1152/ajpheart.1994.267.5.H1984. 400. Clemo HF, Stambler BS, Baumgarten CM. Persistent activation of a
swelling-activated cation current in ventricular myocytes from dogs
385. Zygmunt AC, Gibbons WR. Calcium-activated chloride current in with tachycardia-induced congestive heart failure. Circ Res 83:
rabbit ventricular myocytes. Circ Res 68: 424–437, 1991. doi:10.1161/ 147–157, 1998. doi:10.1161/01.res.83.2.147.
01.res.68.2.424.
401. Clemo HF, Stambler BS, Baumgarten CM. Swelling-activated chlo-
386. Zygmunt AC, Goodrow RJ, Weigel CM. INaCa and ICl(Ca) contrib- ride current is persistently activated in ventricular myocytes from
ute to isoproterenol-induced delayed after depolarizations in mid- dogs with tachycardia-induced congestive heart failure. Circ Res
myocardial cells. Am J Physiol Heart Circ Physiol 275: H1979– 84: 157–165, 1999. doi:10.1161/01.res.84.2.157.
H1992, 1998. doi:10.1152/ajpheart.1998.275.6.H1979.
402. Zou Y, Akazawa H, Qin Y, Sano M, Takano H, Minamino T, Makita
387. Wu MM, Lou J, Song BL, Gong YF, Li YC, Yu CJ, Wang QS, Ma TX, N, Iwanaga K, Zhu W, Kudoh S, Toko H, Tamura K, Kihara M, Nagai
Ma K, Hartzell HC, Duan DD, Zhao D, Zhang ZR. Hypoxia augments T, Fukamizu A, Umemura S, Iiri T, Fujita T, Komuro I. Mechanical
the calcium-activated chloride current carried by anoctamin-1 in stress activates angiotensin II type 1 receptor without the involve-
cardiac vascular endothelial cells of neonatal mice. Br J Pharmacol ment of angiotensin II. Nat Cell Biol 6: 499–506, 2004. doi:10.
171: 3680–3692, 2014. doi:10.1111/bph.12730. 1038/ncb1137.
388. Aggarwal R, Pu J, Boyden PA. Ca21-dependent outward currents 403. Du XY, Sorota S. Cardiac swelling-induced chloride current depo-
in myocytes from epicardial border zone of 5-day infarcted canine larizes canine atrial myocytes. Am J Physiol Heart Circ Physiol
heart. Am J Physiol Heart Circ Physiol 273: H1386–H1394, 1997. 272: H1904–H1916, 1997. doi:10.1152/ajpheart.1997.272.4.H1904.
doi:10.1152/ajpheart.1997.273.3.H1386.
404. Hagiwara N, Masuda H, Shoda M, Irisawa H. Stretch-activated
389. Bahinski A, Nairn AC, Greengard P, Gadsby DC. Chloride conduct- anion currents of rabbit cardiac myocytes. J Physiol 456: 285–302,
ance regulated by cyclic AMP-dependent protein kinase in cardiac 1992. doi:10.1113/jphysiol.1992.sp019337.
myocytes. Nature 340: 718–721, 1989. doi:10.1038/340718a0.
405. Duan DD. The ClC-3 chloride channels in cardiovascular disease.
390. Harvey RD, Hume JR. Autonomic regulation of a chloride current in Acta Pharmacol Sin 32: 675–684, 2011. doi:10.1038/aps.2011.30.
heart. Science 244: 983–985, 1989. doi:10.1126/science.2543073.
406. Decher N, Maier M, Dittrich W, Gassenhuber J, Br€ uggemann A,
391. Harvey RD, Jurevicius JA, Hume JR. Intracellular Na1 modulates Busch AE, Steinmeyer K. Characterization of TASK-4, a novel mem-
the cAMP-dependent regulation of ion channels in the heart. Proc ber of the pH-sensitive, two-pore domain potassium channel family.
Natl Acad Sci USA 88: 6946–6950, 1991. doi:10.1073/pnas.88. FEBS Lett 492: 84–89, 2001. doi:10.1016/s0014-5793(01)02222-0.
16.6946.
407. Lesage F, Guillemare E, Fink M, Duprat F, Lazdunski M, Romey G,
392. Xiang SY, Ye LL, Duan LL, Liu LH, Ge ZD, Auchampach JA, Gross Barhanin J. TWIK-1, a ubiquitous human weakly inward rectifying
GJ, Duan DD. Characterization of a critical role for CFTR chloride K1 channel with a novel structure. EMBO J 15: 1004–1011, 1996.
channels in cardioprotection against ischemia/reperfusion injury.
Acta Pharmacol Sin 32: 824–833, 2011. doi:10.1038/aps.2011.61. 408. O’Connell AD, Morton MJ, Hunter M. Two-pore domain K1 chan-
nels-molecular sensors. Biochim Biophys Acta 1566: 152–161,
393. Tseng GN. Cell swelling increases membrane conductance of ca- 2002. doi:10.1016/S0005-2736(02)00597-7.
nine cardiac cells: evidence for a volume-sensitive Cl channel. Am
J Physiol Cell Physiol 262: C1056–C1068, 1992. doi:10.1152/ 409. Christensen AH, Chatelain FC, Huttner IG, Olesen MS, Soka M,
ajpcell.1992.262.4.C1056. Feliciangeli S, Horvat C, Santiago CF, Vandenberg JI, Schmitt N,

1150 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

Olesen SP, Lesage F, Fatkin D. The two-pore domain potassium cause ventricular tachycardia. EMBO Mol Med 9: 403–414, 2017.
channel, TWIK-1, has a role in the regulation of heart rate and atrial doi:10.15252/emmm.201606690.
size. J Mol Cell Cardiol 97: 24–35, 2016. doi:10.1016/j.yjmcc.2016.
04.006. 422. Lugenbiel P, Wenz F, Syren P, Geschwill P, Govorov K, Seyler C,
Frank D, Schweizer PA, Franke J, Weis T, Bruehl C, Schmack B,
410. Ma L, Zhang X, Zhou M, Chen H. Acid-sensitive TWIK and TASK Ruhparwar A, Karck M, Frey N, Katus HA, Thomas D. TREK-1
two-pore domain potassium channels change ion selectivity and (K2P2.1) K1 channels are suppressed in patients with atrial fibrilla-
become permeable to sodium in extracellular acidification. J Biol tion and heart failure and provide therapeutic targets for rhythm
Chem 287: 37145–37153, 2012. doi:10.1074/jbc.M112.398164. control. Basic Res Cardiol 112: 8, 2017. doi:10.1007/s00395-016-
0597-7.
411. Chen H, Chatelain FC, Lesage F. Altered and dynamic ion selectiv-
ity of K1 channels in cell development and excitability. Trends 423. Schmidt C, Wiedmann F, Schweizer PA, Katus HA, Thomas D.
Pharmacol Sci 35: 461–469, 2014. doi:10.1016/j.tips.2014.06.002. Inhibition of cardiac two-pore-domain K1 (K2P) channels—an
emerging antiarrhythmic concept. Eur J Pharmacol 738: 250–255,
412. Limberg SH, Netter MF, Rolfes C, Rinne S, Schlichtho
€ rl G, Zuzarte 2014. doi:10.1016/j.ejphar.2014.05.056.
M, Vassiliou T, Moosdorf R, Wulf H, Daut J, Sachse FB, Decher N.
TASK-1 channels may modulate action potential duration of human 424. Schmidt C, Wiedmann F, Tristram F, Anand P, Wenzel W, Lugenbiel
atrial cardiomyocytes. Cell Physiol Biochem 28: 613–624, 2011. P, Schweizer PA, Katus HA, Thomas D. Cardiac expression and
doi:10.1159/000335757. atrial fibrillation-associated remodeling of K2p2.1 (TREK-1) K1 chan-
nels in a porcine model. Life Sci 97: 107–115, 2014. doi:10.1016/j.
413. Schmidt C, Wiedmann F, Voigt N, Zhou XB, Heijman J, Lang S, lfs.2013.12.006.
Albert V, Kallenberger S, Ruhparwar A, Szabo  G, Kallenbach K,
Karck M, Borggrefe M, Biliczki P, Ehrlich JR, Baczko  I, Lugenbiel P, 425. Schmidt C, Wiedmann F, Zhou XB, Heijman J, Voigt N, Ratte A,
Schweizer PA, Donner BC, Katus HA, Dobrev D, Thomas D. Lang S, Kallenberger SM, Campana C, Weymann A, De Simone R,
Upregulation of K2P3.1 K1 current causes action potential shorten- Szabo G, Ruhparwar A, Kallenbach K, Karck M, Ehrlich JR, Baczko  I,
ing in patients with chronic atrial fibrillation. Circulation 132: 82–92, Borggrefe M, Ravens U, Dobrev D, Katus HA, Thomas D. Inverse
2015. doi:10.1161/CIRCULATIONAHA.114.012657. remodelling of K2P3.1 K1 channel expression and action potential
duration in left ventricular dysfunction and atrial fibrillation: implica-
414. Schmidt C, Wiedmann F, Kallenberger SM, Ratte A, Schulte JS, tions for patient-specific antiarrhythmic drug therapy. Eur Heart J
Scholz B, M€uller FU, Voigt N, Zafeiriou MP, Ehrlich JR, Tochtermann 38: 1764–1774, 2017. doi:10.1093/eurheartj/ehw559.
U, Veres G, Ruhparwar A, Karck M, Katus HA, Thomas D. Stretch-
activated two-pore-domain (K2P) potassium channels in the heart: 426. Abraham DM, Lee TE, Watson LJ, Mao L, Chandok G, Wang HG,
focus on atrial fibrillation and heart failure. Prog Biophys Mol Biol Frangakis S, Pitt GS, Shah SH, Wolf MJ, Rockman HA. The two-
130: 233–243, 2017. doi:10.1016/j.pbiomolbio.2017.05.004. pore domain potassium channel TREK-1 mediates cardiac fibrosis
and diastolic dysfunction. J Clin Invest 128: 4843–4855, 2018.
415. Wiedmann F, Schulte JS, Gomes B, Zafeiriou MP, Ratte A, Rathjens doi:10.1172/JCI95945.
F, Fehrmann E, Scholz B, Voigt N, M€ uller FU, Thomas D, Katus HA,
Schmidt C. Atrial fibrillation and heart failure-associated remodeling 427. Goonetilleke L, Quayle J. TREK-1 K1 channels in the cardiovascular
of two-pore-domain potassium (K2P) channels in murine disease system: their significance and potential as a therapeutic target.
Cardiovasc Ther 30: e23–e29, 2012. doi:10.1111/j.1755-5922.2010.
models: focus on TASK-1. Basic Res Cardiol 113: 27, 2018.
00227.x.
doi:10.1007/s00395-018-0687-9.

 S, Kiper AK, Schlichtho


€ rl G, Dittmann S, Netter MF, Limberg 428. Schindler RF, Poon KL, Simrick S, Brand T. The Popeye domain
416. Rinne
containing genes: essential elements in heart rate control.
SH, Silbernagel N, Zuzarte M, Moosdorf R, Wulf H, Schulze-Bahr E,
Cardiovasc Diagn Ther 2: 308–319, 2012. doi:10.3978/j.issn.2223-
Rolfes C, Decher N. TASK-1 and TASK-3 may form heterodimers in
3652.2012.12.01.
human atrial cardiomyocytes. J Mol Cell Cardiol 81: 71–80, 2015.
doi:10.1016/j.yjmcc.2015.01.017. 429. Zuo D, Chen K, Zhou M, Liu Z, Chen H. Kir2.1 and K2P1 channels
reconstitute two levels of resting membrane potential in cardiomy-
 S, Kiper AK, Schmidt C, Ortiz-Bonnin B, Zwiener S, Seebohm
417. Rinne
ocytes. J Physiol 595: 5129–5142, 2017. doi:10.1113/JP274268.
G, Decher N. Stress-kinase regulation of TASK-1 and TASK-3. Cell
Physiol Biochem 44: 1024–1037, 2017. doi:10.1159/000485402. 430. Masuda M, de Magalhães Engel G, Barbosa Moreira A.
Characterization of isolated ventricular myocytes: two levels of rest-
 S, Zumhagen S, Kiper AK, Silbernagel N, Netter
418. Friedrich C, Rinne
ing potential. J Mol Cell Cardiol 19: 831–839, 1987. doi:10.1016/
MF, Stallmeyer B, Schulze-Bahr E, Decher N. Gain-of-function muta-
S0022-2828(87)80612-0.
tion in TASK-4 channels and severe cardiac conduction disorder.
EMBO Mol Med 6: 937–951, 2014. doi:10.15252/emmm.20130 431. Wiggins JR, Cranefield PF. Two levels of resting potential in canine
3783. cardiac Purkinje fibers exposed to sodium-free solutions. Circ Res
39: 466–474, 1976. doi:10.1161/01.res.39.4.466.
€ rl G, Daut J. The potassium current carried by
419. Bodnár M, Schlichtho
TREK-1 channels in rat cardiac ventricular muscle. Pflugers Arch 432. Gadsby DC, Cranefield PF. Two levels of resting potential in cardiac
467: 1069–1079, 2015. doi:10.1007/s00424-014-1678-9. Purkinje fibers. J Gen Physiol 70: 725–746, 1977. doi:10.1085/
jgp.70.6.725.
420. Decher N, Kiper AK, Rinne S. Stretch-activated potassium currents
in the heart: Focus on TREK-1 and arrhythmias. Prog Biophys Mol 433. McCullough JR, Chua WT, Rasmussen HH, Ten Eick RE, Singer DH.
Biol 130: 223–232, 2017. doi:10.1016/j.pbiomolbio.2017.05.005. Two stable levels of diastolic potential at physiological K1 concen-
trations in human ventricular myocardial cells. Circ Res 66: 191–
421. Decher N, Ortiz-Bonnin B, Friedrich C, Schewe M, Kiper AK, Rinne  201, 1990. doi:10.1161/01.res.66.1.191.
S, Seemann G, Peyronnet R, Zumhagen S, Bustos D, Kockska €mper
J, Kohl P, Just S, González W, Baukrowitz T, Stallmeyer B, Schulze- 434. Morotti S, McCulloch AD, Bers DM, Edwards AG, Grandi E. Atrial-
Bahr E. Sodium permeable and “hypersensitive” TREK-1 channels selective targeting of arrhythmogenic phase-3 early after-

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1151


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

depolarizations in human myocytes. J Mol Cell Cardiol 96: 63–71, myocytes. Am J Physiol Cell Physiol 278: C546–C553, 2000.
2016. doi:10.1016/j.yjmcc.2015.07.030. doi:10.1152/ajpcell.2000.278.3.C546.

435. Bueno-Orovio A, Sánchez C, Pueyo E, Rodriguez B. Na/K pump 450. Geering K. Functional roles of Na,K-ATPase subunits. Curr Opin
regulation of cardiac repolarization: insights from a systems biology Nephrol Hypertens 17: 526–532, 2008. doi:10.1097/MNH.
approach. Pflugers Arch 466: 183–193, 2014. doi:10.1007/s00424- 0b013e3283036cbf.
013-1293-1.
451. Pavlovic D, Fuller W, Shattock MJ. Novel regulation of cardiac Na
436. Britton OJ, Bueno-Orovio A, Virág L, Varro  A, Rodriguez B. The pump via phospholemman. J Mol Cell Cardiol 61: 83–93, 2013.
electrogenic Na1/K1 pump is a key determinant of repolarization doi:10.1016/j.yjmcc.2013.05.002.
abnormality susceptibility in human ventricular cardiomyocytes: a
population-based simulation study. Front Physiol 8: 278, 2017. 452. Schatzmann HJ. [Cardiac glycosides as inhibitors of active potas-
doi:10.3389/fphys.2017.00278. sium and sodium transport by erythrocyte membrane]. Helv
Physiol Pharmacol Acta 11: 346–354, 1953.
437. Bers DM, Despa S. Na/K-ATPase—an integral player in the adrener-
gic fight-or-flight response. Trends Cardiovasc Med 19: 111–118, 453. Glitsch HG. Electrophysiology of the sodium-potassium-ATPase in
2009. doi:10.1016/j.tcm.2009.07.001. cardiac cells. Physiol Rev 81: 1791–1826, 2001. doi:10.1152/
physrev.2001.81.4.1791.
438. Stein WD. Energetics and the design principles of the Na/K-ATPase. J
Theor Biol 147: 145–159, 1990. doi:10.1016/s0022-5193(05)80049-9. 454. Lytton J. Na1/Ca21 exchangers: three mammalian gene families
control Ca21 transport. Biochem J 406: 365–382, 2007. doi:10.
439. Lingrel J, Moseley A, Dostanic I, Cougnon M, He S, James P, Woo 1042/BJ20070619.
A, O’Connor K, Neumann J. Functional roles of the alpha isoforms
of the Na,K-ATPase. Ann NY Acad Sci 986: 354–359, 2003. doi: 455. Lee SL, Yu AS, Lytton J. Tissue-specific expression of Na1-Ca21
10.1111/j.1749-6632.2003.tb07214.x. exchanger isoforms. J Biol Chem 269: 14849–14852, 1994. doi:
10.1016/S0021-9258(17)36540-7.
440. Shattock MJ, Ottolia M, Bers DM, Blaustein MP, Boguslavskyi A,
Bossuyt J, Bridge JH, Chen-Izu Y, Clancy CE, Edwards A, 456. Nicoll DA, Longoni S, Philipson KD. Molecular cloning and func-
Goldhaber J, Kaplan J, Lingrel JB, Pavlovic D, Philipson K, Sipido tional expression of the cardiac sarcolemmal Na1-Ca21 exchanger.
KR, Xie ZJ. Na1/Ca21 exchange and Na1/K1-ATPase in the heart. Science 250: 562–565, 1990. doi:10.1126/science.1700476.
J Physiol 593: 1361–1382, 2015. doi:10.1113/jphysiol.2014.282319.
457. Nicoll DA, Quednau BD, Qui Z, Xia YR, Lusis AJ, Philipson KD.
441. Despa S, Bers DM. Functional analysis of Na1/K1-ATPase isoform Cloning of a third mammalian Na1-Ca21 exchanger, NCX3. J Biol
distribution in rat ventricular myocytes. Am J Physiol Cell Physiol Chem 271: 24914–24921, 1996. doi:10.1074/jbc.271.40.24914.
293: C321–C327, 2007. doi:10.1152/ajpcell.00597.2006.
458. Quednau BD, Nicoll DA, Philipson KD. Tissue specificity and alter-
442. Despa S, Lingrel JB, Bers DM. Na1/K1-ATPase alpha2-isoform native splicing of the Na1/Ca21 exchanger isoforms NCX1, NCX2,
preferentially modulates Ca21 transients and sarcoplasmic reticu- and NCX3 in rat. Am J Physiol Cell Physiol 272: C1250–C1261,
lum Ca21 release in cardiac myocytes. Cardiovasc Res 95: 480– 1997. doi:10.1152/ajpcell.1997.272.4.C1250.
486, 2012. doi:10.1093/cvr/cvs213.
459. Ren X, Philipson KD. The topology of the cardiac Na1/Ca21
443. Yuen GK, Galice S, Bers DM. Subcellular localization of Na/K- exchanger, NCX1. J Mol Cell Cardiol 57: 68–71, 2013. doi:10.1016/j.
ATPase isoforms in ventricular myocytes. J Mol Cell Cardiol 108: yjmcc.2013.01.010.
158–169, 2017. doi:10.1016/j.yjmcc.2017.05.013.
460. Khananshvili D. The SLC8 gene family of sodium-calcium exchang-
444. Berry RG, Despa S, Fuller W, Bers DM, Shattock MJ. Differential dis- ers (NCX)—structure, function, and regulation in health and dis-
tribution and regulation of mouse cardiac Na1/K1-ATPase alpha1 ease. Mol Aspects Med 34: 220–235, 2013. doi:10.1016/j.mam.
and alpha2 subunits in T-tubule and surface sarcolemmal mem- 2012.07.003.
branes. Cardiovasc Res 73: 92–100, 2007. doi:10.1016/j.cardiores.
2006.11.006. 461. Matsuoka S, Nicoll DA, He Z, Philipson KD. Regulation of cardiac
Na1-Ca21 exchanger by the endogenous XIP region. J Gen
445. Eisner DA, Lederer WJ, Vaughan-Jones RD. The electrogenic Na Physiol 109: 273–286, 1997. doi:10.1085/jgp.109.2.273.
pump in mammalian cardiac muscle. Soc Gen Physiol Ser 38: 193–
213, 1984. 462. Szerencsei RT, Kinjo TG, Schnetkamp PP. The topology of the C-
terminal sections of the NCX1 Na1/Ca21 exchanger and the
446. Bossuyt J, Despa S, Han F, Hou Z, Robia SL, Lingrel JB, Bers DM. NCKX2 Na1/Ca21-K1 exchanger. Channels (Austin) 7: 109–114,
Isoform specificity of the Na/K-ATPase association and regulation 2013. doi:10.4161/chan.23898.
by phospholemman. J Biol Chem 284: 26749–26757, 2009.
doi:10.1074/jbc.M109.047357. 463. Eisner DA, Caldwell JL, Kistamás K, Trafford AW. Calcium and exci-
tation-contraction coupling in the heart. Circ Res 121: 181–195, 2017.
447. Glitsch HG, Krahn T, Pusch H, Suleymanian M. Effect of isoprena- doi:10.1161/CIRCRESAHA.117.310230.
line on active Na transport in sheep cardiac Purkinje fibres.
Pflugers Arch 415: 88–94, 1989. doi:10.1007/BF00373145. 464. Hurtado C, Prociuk M, Maddaford TG, Dibrov E, Mesaeli N, Hryshko
LV, Pierce GN. Cells expressing unique Na1/Ca21 exchange
448. Shah A, Cohen IS, Rosen MR. Stimulation of cardiac alpha recep- (NCX1) splice variants exhibit different susceptibilities to Ca21 over-
tors increases Na/K pump current and decreases gK via a pertussis load. Am J Physiol Heart Circ Physiol 290: H2155–H2162, 2006.
toxin-sensitive pathway. Biophys J 54: 219–225, 1988. doi:10.1016/ doi:10.1152/ajpheart.00958.2005.
S0006-3495(88)82950-3.
465. Kraev A, Chumakov I, Carafoli E. Molecular biological studies of the
449. Hansen PS, Buhagiar KA, Gray DF, Rasmussen HH. Voltage-de- cardiac sodium-calcium exchanger. Ann NY Acad Sci 779: 103–
pendent stimulation of the Na1-K1 pump by insulin in rabbit cardiac 109, 1996. doi:10.1111/j.1749-6632.1996.tb44774.x.

1152 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

466. Li GR, Nattel S. Demonstration of an inward Na1-Ca21 exchange 478. Bovo E, de Tombe PP, Zima AV. The role of dyadic organization in
current in adult human atrial myocytes. Ann NY Acad Sci 779: regulation of sarcoplasmic reticulum Ca21 handling during rest in
525–528, 1996. doi:10.1111/j.1749-6632.1996.tb44827.x. rabbit ventricular myocytes. Biophys J 106: 1902–1909, 2014.
doi:10.1016/j.bpj.2014.03.032.
467. Sher AA, Noble PJ, Hinch R, Gavaghan DJ, Noble D. The role of
the Na1/Ca21 exchangers in Ca21 dynamics in ventricular myo- 479. Garciarena CD, Youm JB, Swietach P, Vaughan-Jones RD. H1-acti-
cytes. Prog Biophys Mol Biol 96: 377–398, 2008. doi:10.1016/j. vated Na1 influx in the ventricular myocyte couples Ca21-signalling
pbiomolbio.2007.07.018. to intracellular pH. J Mol Cell Cardiol 61: 51–59, 2013. doi:10.1016/j.
yjmcc.2013.04.008.
468. Bers DM, Weber CR. Na/Ca exchange function in intact ventricular
myocytes. Ann NY Acad Sci 976: 500–512, 2002. doi:10.1111/j.1749- 480. Sipido KR, Acsai K, Antoons G, Bito V, Macquaide N. T-tubule
6632.2002.tb04784.x. remodelling and ryanodine receptor organization modulate so-
dium-calcium exchange. Adv Exp Med Biol 961: 375–383, 2013.
469. Kohajda Z, Farkas-Morvay N, Jost N, Nagy N, Geramipour A, doi:10.1007/978-1-4614-4756-6_32.
Horvath A, Varga RS, Hornyik T, Corici C, Acsai K, Horváth B,
481. Scriven DR, Moore ED. Ca21 channel and Na1/Ca21 exchange
Prorok J, Ördo € g B, De  ri S, To  th D, Levijoki J, Pollesello P,
localization in cardiac myocytes. J Mol Cell Cardiol 58: 22–31,
Koskelainen T, Otsomaa L, To  th A, Baczko I, Leprán I, Nánási PP,
2013. doi:10.1016/j.yjmcc.2012.11.022.
Papp JG, Varro  A, Virág L. The effect of a novel highly selective in-
hibitor of the sodium/calcium exchanger (NCX) on cardiac arrhyth- 482. Levesque PC, Leblanc N, Hume JR. Role of reverse-mode Na1-
mias in in vitro and in vivo experiments. PLoS One 11: e0166041, Ca21 exchange in excitation-contraction coupling in the heart. Ann
2016. doi:10.1371/journal.pone.0166041. NY Acad Sci 639: 386–397, 1991. doi:10.1111/j.1749-6632.1991.
tb17327.x.
470. Nagy N, Kormos A, Kohajda Z, Szebeni A, Szepesi J, Pollesello P,
 A, To
Levijoki J, Acsai K, Virág L, Nánási PP, Papp JG, Varro  th A. 483. Acsai K, Antoons G, Livshitz L, Rudy Y, Sipido KR. Microdomain
Selective Na1/Ca21 exchanger inhibition prevents Ca21 overload- [Ca21] near ryanodine receptors as reported by L-type Ca21 and
induced triggered arrhythmias. Br J Pharmacol 171: 5665–5681, Na1/Ca21 exchange currents. J Physiol 589: 2569–2583, 2011.
2014. doi:10.1111/bph.12867. doi:10.1113/jphysiol.2010.202663.

471. Christ T, Kovács PP, Acsai K, Knaut M, Eschenhagen T, Jost N, 484. Zamparelli C, Macquaide N, Colotti G, Verzili D, Seidler T, Smith GL,
 A, Wettwer E, Ravens U. Block of Na1/Ca21 exchanger by
Varro Chiancone E. Activation of the cardiac Na1-Ca21 exchanger by
SEA0400 in human right atrial preparations from patients in sinus sorcin via the interaction of the respective Ca21-binding domains. J
rhythm and in atrial fibrillation. Eur J Pharmacol 788: 286–293, Mol Cell Cardiol 49: 132–141, 2010. doi:10.1016/j.yjmcc.2010.03.
2016. doi:10.1016/j.ejphar.2016.06.050. 003.

472. Hurtado C, Ander BP, Maddaford TG, Lukas A, Hryshko LV, Pierce 485. Villa-Abrille MC, Sidor A, O’Rourke B. Insulin effects on cardiac
GN. Adenovirally delivered shRNA strongly inhibits Na1-Ca21 Na1/Ca21 exchanger activity: role of the cytoplasmic regulatory
loop. J Biol Chem 283: 16505–16513, 2008. doi:10.1074/jbc.
exchanger expression but does not prevent contraction of neona-
M801424200.
tal cardiomyocytes. J Mol Cell Cardiol 38: 647–654, 2005. doi:10.
1016/j.yjmcc.2005.02.007. 486. Condrescu M, Reeves JP. Actin-dependent regulation of the car-
diac Na1/Ca21 exchanger. Am J Physiol Cell Physiol 290: C691–
473. Puglisi JL, Bassani RA, Bassani JW, Amin JN, Bers DM.
C701, 2006. doi:10.1152/ajpcell.00232.2005.
Temperature and relative contributions of Ca transport systems in
cardiac myocyte relaxation. Am J Physiol Heart Circ Physiol 270: 487. Whitmer JT, Idell-Wenger JA, Rovetto MJ, Neely JR. Control of fatty
H1772–H1778, 1996. doi:10.1152/ajpheart.1996.270.5.H1772. acid metabolism in ischemic and hypoxic hearts. J Biol Chem 253:
4305–4309, 1978.
474. Jost N, Nagy N, Corici C, Kohajda Z, Horváth A, Acsai K, Biliczki P,
 th A, Papp JG,
Levijoki J, Pollesello P, Koskelainen T, Otsomaa L, To 488. Finck BN, Han X, Courtois M, Aimond F, Nerbonne JM, Kovacs A,
Varro A, Virág L. ORM-10103, a novel specific inhibitor of the Na1/ Gross RW, Kelly DP. A critical role for PPARalpha-mediated lipotox-
Ca21 exchanger, decreases early and delayed afterdepolarizations icity in the pathogenesis of diabetic cardiomyopathy: modulation
in the canine heart. Br J Pharmacol 170: 768–778, 2013. doi:10.1111/ by dietary fat content. Proc Natl Acad Sci USA 100: 1226–1231,
bph.12228. 2003. doi:10.1073/pnas.0336724100.

475. Nagy ZA, Virág L, To th A, Biliczki P, Acsai K, Bányász T, Nánási P, 489. Sharma S, Adrogue JV, Golfman L, Uray I, Lemm J, Youker K, Noon
Papp JG, Varro A. Selective inhibition of sodium-calcium exchanger GP, Frazier OH, Taegtmeyer H. Intramyocardial lipid accumulation
by SEA-0400 decreases early and delayed after depolarization in in the failing human heart resembles the lipotoxic rat heart. FASEB
canine heart. Br J Pharmacol 143: 827–831, 2004. doi:10.1038/sj. J 18: 1692–1700, 2004. doi:10.1096/fj.04-2263com.
bjp.0706026. 490. Riedel MJ, Baczko  I, Searle GJ, Webster N, Fercho M, Jones L,
476. Zhao Z, Wen H, Fefelova N, Allen C, Baba A, Matsuda T, Xie LH. Lang J, Lytton J, Dyck JR, Light PE. Metabolic regulation of sodium-
calcium exchange by intracellular acyl CoAs. EMBO J 25: 4605–
Revisiting the ionic mechanisms of early afterdepolarizations in car-
4614, 2006. doi:10.1038/sj.emboj.7601321.
diomyocytes: predominant by Ca waves or Ca currents? Am J
Physiol Heart Circ Physiol 302: H1636–H1644, 2012. doi:10.1152/ 491. Orchard CH, Cingolani HE. Acidosis and arrhythmias in cardiac
ajpheart.00742.2011. muscle. Cardiovasc Res 28: 1312–1319, 1994. doi:10.1093/cvr/
28.9.1312.
477. Milberg P, Pott C, Fink M, Frommeyer G, Matsuda T, Baba A, Osada
N, Breithardt G, Noble D, Eckardt L. Inhibition of the Na1/Ca21 492. Scranton K, John S, Escobar A, Goldhaber J, Ottolia M. Modulation
exchanger suppresses torsades de pointes in an intact heart model of the cardiac Na1-Ca21 exchanger by cytoplasmic protons: molec-
of long QT syndrome-2 and long QT syndrome-3. Heart Rhythm 5: ular mechanisms and physiological implications. Cell Calcium 87:
1444–1452, 2008. doi:10.1016/j.hrthm.2008.06.017. 102140, 2020. doi:10.1016/j.ceca.2019.102140.

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1153


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

493. Langenbacher AD, Dong Y, Shu X, Choi J, Nicoll DA, Goldhaber JI, 509. Despa S. Myocyte [Na1]i dysregulation in heart failure and diabetic
Philipson KD, Chen JN. Mutation in sodium-calcium exchanger 1 cardiomyopathy. Front Physiol 9: 1303, 2018. doi:10.3389/fphys.
(NCX1) causes cardiac fibrillation in zebrafish. Proc Natl Acad Sci 2018.01303.
USA 102: 17699–17704, 2005. doi:10.1073/pnas.0502679102.
510. Frelin C, Vigne P, Lazdunski M. The role of the Na1/H1
494. Murer H, Hopfer U, Kinne R. Sodium/proton antiport in brush-bor- exchange system in the regulation of the internal pH in cultured
der-membrane vesicles isolated from rat small intestine and kidney. cardiac cells. Eur J Biochem 149: 1–4, 1985. doi:10.1111/j.1432-
Biochem J 154: 597–604, 1976. doi:10.1042/bj1540597. 1033.1985.tb08884.x.

495. Orlowski J, Grinstein S. Na1/H1 exchangers of mammalian cells. J 511. Baczko  I, Mraiche F, Light PE, Fliegel L. Diastolic calcium is elevated
Biol Chem 272: 22373–22376, 1997. doi:10.1074/jbc.272.36. in metabolic recovery of cardiomyocytes expressing elevated lev-
22373. els of the Na1/H1 exchanger. Can J Physiol Pharmacol 86: 850–
859, 2008. doi:10.1139/Y08-092.
496. Wakabayashi S, Hisamitsu T, Nakamura TY. Regulation of the car-
512. Bkaily G, Chahine M, Al-Khoury J, Avedanian L, Beier N, Scholz W,
diac Na1/H1 exchanger in health and disease. J Mol Cell Cardiol
Jacques D. Na1-H1 exchanger inhibitor prevents early death in he-
61: 68–76, 2013. doi:10.1016/j.yjmcc.2013.02.007.
reditary cardiomyopathy. Can J Physiol Pharmacol 93: 923–934,
497. Fliegel L, Dyck JR, Wang H, Fong C, Haworth RS. Cloning and anal- 2015. doi:10.1139/cjpp-2015-0107.
ysis of the human myocardial Na1/H1 exchanger. Mol Cell
513. Bkaily G, Jacques D. Na1-H1 exchanger and proton channel in
Biochem 125: 137–143, 1993. doi:10.1007/BF00936442.
heart failure associated with Becker and Duchenne muscular dys-
498. Aharonovitz O, Zaun HC, Balla T, York JD, Orlowski J, Grinstein S. trophies. Can J Physiol Pharmacol 95: 1213–1223, 2017. doi:10.
Intracellular pH regulation by Na1/H1 exchange requires phospha- 1139/cjpp-2017-0265.
tidylinositol 4,5-bisphosphate. J Cell Biol 150: 213–224, 2000. 514. Packer M. Activation and inhibition of sodium-hydrogen exchanger
doi:10.1083/jcb.150.1.213. is a mechanism that links the pathophysiology and treatment of dia-
betes mellitus with that of heart failure. Circulation 136: 1548–1559,
€ ster S, Pavkov-Keller T, K€
499. Ko uhlbrandt W, Yildiz O. Structure of
2017. doi:10.1161/CIRCULATIONAHA.117.030418.
human Na1/H1 exchanger NHE1 regulatory region in complex with
calmodulin and Ca21. J Biol Chem 286: 40954–40961, 2011. 515. DiFrancesco D. The role of the funny current in pacemaker activ-
doi:10.1074/jbc.M111.286906. ity. Circ Res 106: 434–446, 2010. doi:10.1161/CIRCRESAHA.109.
208041.
500. Lee BL, Sykes BD, Fliegel L. Structural and functional insights into
the cardiac Na1/H1 exchanger. J Mol Cell Cardiol 61: 60–67, 2013. 516. DiFrancesco D, Noble D. Rebuttal: “The funny current in the con-
doi:10.1016/j.yjmcc.2012.11.019. text of the coupled clock pacemaker cell system”. Heart Rhythm 9:
457–458, 2012. doi:10.1016/j.hrthm.2011.09.023.
501. Orlowski J, Kandasamy RA. Delineation of transmembrane
domains of the Na1/H1 exchanger that confer sensitivity to phar- 517. Morad M, Zhang XH. Mechanisms of spontaneous pacing: sinoa-
macological antagonists. J Biol Chem 271: 19922–19927, 1996. trial nodal cells, neonatal cardiomyocytes, and human stem cell
doi:10.1074/jbc.271.33.19922. derived cardiomyocytes. Can J Physiol Pharmacol 95: 1100–1107,
2017. doi:10.1139/cjpp-2016-0743.
502. Pang T, Su X, Wakabayashi S, Shigekawa M. Calcineurin homolo-
gous protein as an essential cofactor for Na1/H1 exchangers. J 518. Verkerk AO, Wilders R, van Borren MM, Peters RJ, Broekhuis E,
Biol Chem 276: 17367–17372, 2001. doi:10.1074/jbc.M100296200. Lam K, Coronel R, de Bakker JM, Tan HL. Pacemaker current (If) in
the human sinoatrial node. Eur Heart J 28: 2472–2478, 2007.
503. Sardet C, Franchi A, Pouysse  gur J. Molecular cloning, primary doi:10.1093/eurheartj/ehm339.
structure, and expression of the human growth factor-activatable
Na1/H1 antiporter. Cell 56: 271–280, 1989. doi:10.1016/0092-8674 519. Vinogradova TM, Tagirova Sirenko S, Lakatta EG. Unique Ca21-cy-
(89)90901-x. cling protein abundance and regulation sustains local Ca21
releases and spontaneous firing of rabbit sinoatrial node cells. Int J
504. Wakabayashi S, Pang T, Su X, Shigekawa M. A novel topology Mol Sci 19: 2173, 2018. doi:10.3390/ijms19082173.
model of the human Na1/H1 exchanger isoform 1. J Biol Chem
520. Weidmann S. Effect of current flow on the membrane potential of
275: 7942–7949, 2000. doi:10.1074/jbc.275.11.7942.
cardiac muscle. J Physiol 115: 227–236, 1951. doi:10.1113/jphysiol.
505. Fafournoux P, Noël J, Pouysse gur J. Evidence that Na1/H1 1951.sp004667.
exchanger isoforms NHE1 and NHE3 exist as stable dimers in mem-
521. Hauswirth O, Noble D, Tsien RW. Adrenaline: mechanism of action
branes with a high degree of specificity for homodimers. J Biol
on the pacemaker potential in cardiac Purkinje fibers. Science 162:
Chem 269: 2589–2596, 1994. 916–917, 1968. doi:10.1126/science.162.3856.916.
506. Fuster D, Moe OW, Hilgemann DW. Steady-state function of the 522. Hauswirth O, Noble D, Tsien RW. Separation of the pace-maker
ubiquitous mammalian Na/H exchanger (NHE1) in relation to dimer and plateau components of delayed rectification in cardiac
coupling models with 2Na/2H stoichiometry. J Gen Physiol 132: Purkinje fibres. J Physiol 225: 211–235, 1972. doi:10.1113/jphysiol.
465–480, 2008. doi:10.1085/jgp.200810016. 1972.sp009934.
507. Wakabayashi S, Shigekawa M, Pouyssegur J. Molecular physiology 523. Alings AM, Bouman LN. Electrophysiology of the ageing rabbit and
of vertebrate Na1/H1 exchangers. Physiol Rev 77: 51–74, 1997. cat sinoatrial node–a comparative study. Eur Heart J 14: 1278–
doi:10.1152/physrev.1997.77.1.51. 1288, 1993. doi:10.1093/eurheartj/14.9.1278.

508. Alexander RT, Grinstein S. Na1/H1 exchangers and the regulation 524. DiFrancesco D. A new interpretation of the pace-maker current in
of volume. Acta Physiol (Oxf) 187: 159–167, 2006. doi:10.1111/j.1748- calf Purkinje fibres. J Physiol 314: 359–376, 1981. doi:10.1113/
1716.2006.01558.x. jphysiol.1981.sp013713.

1154 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

525. Maylie J, Morad M. Ionic currents responsible for the generation of 540. Ono K, Shibata S, Iijima T. Properties of the delayed rectifier potas-
pace-maker current in the rabbit sino-atrial node. J Physiol 355: sium current in porcine sino-atrial node cells. J Physiol 524: 51–62,
215–235, 1984. doi:10.1113/jphysiol.1984.sp015415. 2000. doi:10.1111/j.1469-7793.2000.00051.x.

526. Hagiwara N, Irisawa H, Kameyama M. Contribution of two types of 541. DiFrancesco D. Funny channel gene mutations associated with
calcium currents to the pacemaker potentials of rabbit sino-atrial arrhythmias. J Physiol 591: 4117–4124, 2013. doi:10.1113/jphysiol.
node cells. J Physiol 395: 233–253, 1988. doi:10.1113/jphysiol.1988. 2013.253765.
sp016916.
542. DiFrancesco D, Mangoni M. Modulation of single hyperpolariza-
527. Li Y, Fu X, Zhang Z, Yu B. Knockdown of cardiac Kir3.1 gene with tion-activated channels (if) by cAMP in the rabbit sino-atrial node. J
siRNA can improve bradycardia in an experimental sinus bradycar- Physiol 474: 473–482, 1994. doi:10.1113/jphysiol.1994.sp020038.
dia rat model. Mol Cell Biochem 429: 103–111, 2017. doi:10.1007/
543. Mesirca P, Marger L, Toyoda F, Rizzetto R, Audoubert M, Dubel S,
s11010-017-2939-7.
Torrente AG, Difrancesco ML, Muller JC, Leoni AL, Couette B,
528. Verkerk AO, van Borren MM, Wilders R. Calcium transient and so- Nargeot J, Clapham DE, Wickman K, Mangoni ME. The G-protein-
dium-calcium exchange current in human versus rabbit sinoatrial gated K1 channel, IKACh, is required for regulation of pacemaker
node pacemaker cells. ScientificWorldJournal 2013: 507872, activity and recovery of resting heart rate after sympathetic stimula-
2013., doi:10.1155/2013/507872. tion. J Gen Physiol 142: 113–126, 2013. doi:10.1085/jgp.201310996.

529. Verkerk AO, Wilders R, van Borren MM, Tan HL. Is sodium current 544. Mangoni ME, Couette B, Bourinet E, Platzer J, Reimer D, Striessnig
present in human sinoatrial node cells? Int J Biol Sci 5: 201–204, J, Nargeot J. Functional role of L-type Cav1.3 Ca21 channels in car-
diac pacemaker activity. Proc Natl Acad Sci USA 100: 5543–5548,
2009. doi:10.7150/ijbs.5.201.
2003. doi:10.1073/pnas.0935295100.
530. Noma A, Morad M, Irisawa H. Does the “pacemaker current” gener-
545. Mangoni ME, Nargeot J. Genesis and regulation of the heart auto-
ate the diastolic depolarization in the rabbit SA node cells?
maticity. Physiol Rev 88: 919–982, 2008. doi:10.1152/physrev.
Pflugers Arch 397: 190–194, 1983. doi:10.1007/BF00584356.
00018.2007.
531. Capel RA, Terrar DA. Cytosolic calcium ions exert a major influence
546. Zaniboni M, Cacciani F, Lux RL. Beat-to-beat cycle length variability
on the firing rate and maintenance of pacemaker activity in guinea-
of spontaneously beating guinea pig sinoatrial cells: relative contri-
pig sinus node. Front Physiol 6: 23, 2015. doi:10.3389/fphys.
butions of the membrane and calcium clocks. PLoS One 9:
2015.00023.
e100242, 2014. doi:10.1371/journal.pone.0100242.
532. Capel RA, Terrar DA. The importance of Ca21-dependent mecha-
547. Baruscotti M, Bianco E, Bucchi A, DiFrancesco D. Current under-
nisms for the initiation of the heartbeat. Front Physiol 6: 80, 2015. standing of the pathophysiological mechanisms responsible for
doi:10.3389/fphys.2015.00080. inappropriate sinus tachycardia: role of the If “funny” current. J
Interv Card Electrophysiol 46: 19–28, 2016. doi:10.1007/s10840-
533. H€user J, Blatter LA, Lipsius SL. Intracellular Ca21 release contrib-
015-0097-y.
utes to automaticity in cat atrial pacemaker cells. J Physiol 524:
415–422, 2000. doi:10.1111/j.1469-7793.2000.00415.x. 548. Huang X, Yang P, Du Y, Zhang J, Ma A. Age-related down-regula-
tion of HCN channels in rat sinoatrial node. Basic Res Cardiol 102:
534. Lakatta EG, Maltsev VA, Bogdanov KY, Stern MD, Vinogradova TM.
429–435, 2007. doi:10.1007/s00395-007-0660-5.
Cyclic variation of intracellular calcium: a critical factor for cardiac
pacemaker cell dominance. Circ Res 92: e45–e50, 2003. doi:10. 549. Caballero R, de la Fuente MG, Go  mez R, Barana A, Amoro  s I, Dolz-
1161/01.res.0000055920.64384.fb. Gaito  n P, Osuna L, Almendral J, Atienza F, Fernandez-Avile s F, Pita
A, Rodríguez-Roda J, Pinto A, Tamargo J, Delpo  n E. In humans,
535. Lakatta EG, DiFrancesco D. What keeps us ticking: a funny current,
chronic atrial fibrillation decreases the transient outward current
a calcium clock, or both? J Mol Cell Cardiol 47: 157–170, 2009.
and ultrarapid component of the delayed rectifier current differen-
doi:10.1016/j.yjmcc.2009.03.022.
tially on each atria and increases the slow component of the
536. Boyett MR, Honjo H, Kodama I. The sinoatrial node, a heterogene- delayed rectifier current in both. J Am Coll Cardiol 55: 2346–2354,
ous pacemaker structure. Cardiovasc Res 47: 658–687, 2000. 2010. doi:10.1016/j.jacc.2010.02.028.
doi:10.1016/s0008-6363(00)00135-8. 550. Sánchez C, Bueno-Orovio A, Wettwer E, Loose S, Simon J, Ravens
chiga-Figueroa IA, Rodríguez-Martinez M, Sánchez-Chapula JA. U, Pueyo E, Rodriguez B. Inter-subject variability in human atrial
537. Are
action potential in sinus rhythm versus chronic atrial fibrillation.
Voltage-dependent potassium currents in feline sino-atrial node
PLoS One 9: e105897, 2014. doi:10.1371/journal.pone.0105897.
myocytes. Pflugers Arch 462: 385–396, 2011. doi:10.1007/s00424-
011-0984-8. 551. Xu ZX, Jin MW. Characterization of transient outward K1 current
and ultra-rapid delayed rectifier K1 current in isolated human atrial
538. Kohajda Z, To th N, Szlovák J, Loewe A, Bitay G, Gazdag P, Prorok
myocytes from patients with congestive heart failure. Acta
 A, Nagy N. Novel
J, Jost N, Levijoki J, Pollesello P, Papp JG, Varro
Pharmacol Sin 23: 110–116, 2002.
Na1/Ca21 exchanger inhibitor ORM-10962 supports coupled func-
tion of funny-current and Na1/Ca21 exchanger in pacemaking of 552. Ravens U, Poulet C, Wettwer E, Knaut M. Atrial selectivity of antiar-
rabbit sinus node tissue. Front Pharmacol 10: 1632, 2019. doi: rhythmic drugs. J Physiol 591: 4087–4097, 2013. doi:10.1113/
10.3389/fphar.2019.01632. jphysiol.2013.256115.

539. Lei M, Cooper PJ, Camelliti P, Kohl P. Role of the 293b-sensitive, 553. Li GR, Feng J, Wang Z, Fermini B, Nattel S. Comparative mecha-
slowly activating delayed rectifier potassium current, IKs, in pace- nisms of 4-aminopyridine-resistant Ito in human and rabbit atrial
maker activity of rabbit isolated sino-atrial node cells. Cardiovasc myocytes. Am J Physiol Heart Circ Physiol 269: H463–H472,
Res 53: 68–79, 2002. doi:10.1016/S0008-6363(01)00459-X. 1995. doi:10.1152/ajpheart.1995.269.2.H463.

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1155


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

554. Li GR, Feng J, Wang Z, Nattel S. Transmembrane chloride currents 567. Brunton TL, Fayrer J. XXII. Note on independent pulsation of the
in human atrial myocytes. Am J Physiol Cell Physiol 270: C500– pulmonary veins and vena cava. Proc R Soc Lond 25: 174–176,
C507, 1996. doi:10.1152/ajpcell.1996.270.2.C500. 1877. doi:10.1098/rspl.1876.0041

555. Xiao GS, Zhang YH, Wang Y, Sun HY, Baumgarten CM, Li GR. 568. Cheung DW. Electrical activity of the pulmonary vein and its interac-
Noradrenaline up-regulates volume-regulated chloride current by tion with the right atrium in the guinea-pig. J Physiol 314: 445–456,
PKA-independent cAMP/exchange protein activated by cAMP 1981. doi:10.1113/jphysiol.1981.sp013718.
pathway in human atrial myocytes. Br J Pharmacol 175: 3422–
569. Haïssaguerre M, Jaïs P, Shah DC, Takahashi A, Hocini M, Quiniou
3432, 2018. doi:10.1111/bph.14392. G, Garrigue S, Le Mouroux A, Le MP, Cle  menty J. Spontaneous ini-
556. Casini S, Marchal GA, Kawasaki M, Nariswari FA, Portero V, van tiation of atrial fibrillation by ectopic beats originating in the pulmo-
nary veins. N Engl J Med 339: 659–666, 1998. doi:10.1056/
den Berg NW, Guan K, Driessen AH, Veldkamp MW, Mengarelli I,
NEJM199809033391003.
de Groot JR, Verkerk AO, Remme CA. Absence of functional
Nav1.8 channels in non-diseased atrial and ventricular cardiomyo- 570. Hocini M, Ho SY, Kawara T, Linnenbank AC, Potse M, Shah D, Jaïs
cytes. Cardiovasc Drugs Ther 33: 649–660, 2019. doi:10.1007/ P, Janse MJ, Haïssaguerre M, De Bakker JM. Electrical conduction
s10557-019-06925-6. in canine pulmonary veins: electrophysiological and anatomic cor-
relation. Circulation 105: 2442–2448, 2002. doi:10.1161/01.cir.
557. Be nitah JP, Go mez AM, Fauconnier J, Kerfant BG, Perrier E, 0000016062.80020.11.
Vassort G, Richard S. Voltage-gated Ca21 currents in the human
pathophysiologic heart: a review. Basic Res Cardiol 97, Suppl 1: 571. Chen YJ, Chen SA. Electrophysiology of pulmonary veins. J
I11–I18, 2002. doi:10.1007/s003950200023. Cardiovasc Electrophysiol 17: 220–224, 2006. doi:10.1111/j.1540-
8167.2005.00317.x.
558. Juhász V, Hornyik T, Benák A, Nagy N, Husti Z, Pap R, Sághy L,
Virág L, Varro A, Baczko
 I. Comparison of the effects of IK,ACh, IKr, 572. Roney CH, Bayer JD, Cochet H, Meo M, Dubois R, Jaïs P, Vigmond
and INa block in conscious dogs with atrial fibrillation and on action EJ. Variability in pulmonary vein electrophysiology and fibrosis
potentials in remodeled atrial trabeculae. Can J Physiol Pharmacol determines arrhythmia susceptibility and dynamics. PLoS Comput
96: 18–25, 2018. doi:10.1139/cjpp-2017-0342. Biol 14: e1006166, 2018. doi:10.1371/journal.pcbi.1006166.

573. Huang SY, Chen YC, Kao YH, Hsieh MH, Chen YA, Chen WP, Lin
559. Oshikawa N, Watanabe I, Masaki R, Shindo A, Kojima T, Saito S,
YK, Chen SA, Chen YJ. Renal failure induces atrial arrhythmogene-
Ozawa Y, Kanmatsuse K. Frequency-dependent electrophysiologi-
sis from discrepant electrophysiological remodeling and calcium
cal effect of ibutilide on human atrium and ventricle. J Interv Card
regulation in pulmonary veins, sinoatrial node, and atria. Int J
Electrophysiol 5: 81–87, 2001. doi:10.1023/a:1009866126492.
Cardiol 202: 846–857, 2016. doi:10.1016/j.ijcard.2015.10.004.
560. Du XL, Lau CP, Chiu SW, Tse HF, Gerlach U, Li GR. Effects of chro- 574. Huang SY, Chen YC, Kao YH, Hsieh MH, Lin YK, Chung CC, Lee TI,
manol 293B on transient outward and ultra-rapid delayed rectifier Tsai WC, Chen SA, Chen YJ. Fibroblast growth factor 23 dysregu-
potassium currents in human atrial myocytes. J Mol Cell Cardiol lates late sodium current and calcium homeostasis with enhanced
35: 293–300, 2003. doi:10.1016/S0022-2828(03)00007-5. arrhythmogenesis in pulmonary vein cardiomyocytes. Oncotarget
7: 69231–69242, 2016. doi:10.18632/oncotarget.12470.
561. Peng G, Barro-Soria R, Sampson KJ, Larsson HP, Kass RS. Gating
mechanisms underlying deactivation slowing by two KCNQ1 atrial 575. Chen YJ, Chen SA, Chen YC, Yeh HI, Chan P, Chang MS, Lin CI.
fibrillation mutations. Sci Rep 7: 45911, 2017. doi:10.1038/srep45911. Effects of rapid atrial pacing on the arrhythmogenic activity of sin-
gle cardiomyocytes from pulmonary veins: implication in initiation
562. Qi XY, Diness JG, Brundel BJ, Zhou XB, Naud P, Wu CT, Huang H, of atrial fibrillation. Circulation 104: 2849–2854, 2001. doi:10.1161/
Harada M, Aflaki M, Dobrev D, Grunnet M, Nattel S. Role of small- hc4801.099736.
conductance calcium-activated potassium channels in atrial elec-
trophysiology and fibrillation in the dog. Circulation 129: 430–440, 576. Ehrlich JR, Cha TJ, Zhang L, Chartier D, Melnyk P, Hohnloser SH,
2014. doi:10.1161/CIRCULATIONAHA.113.003019. Nattel S. Cellular electrophysiology of canine pulmonary vein cardi-
omyocytes: action potential and ionic current properties. J Physiol
563. Davis LD. Effects of autonomic neurohumors on transmembrane 551: 801–813, 2003. doi:10.1113/jphysiol.2003.046417.
potentials of atrial plateau fibers. Am J Physiol 229: 1351–1356,
1975. doi:10.1152/ajplegacy.1975.229.5.1351. 577. Hamaguchi S, Hikita K, Tanaka Y, Tsuneoka Y, Namekata I, Tanaka
H. Enhancement of automaticity by mechanical stretch of the iso-
564. Li N, Csepe TA, Hansen BJ, Sul LV, Kalyanasundaram A, Zakharkin lated guinea pig pulmonary vein myocardium. Biol Pharm Bull 39:
SO, Zhao J, Guha A, Van Wagoner DR, Kilic A, Mohler PJ, Janssen 1216–1219, 2016. doi:10.1248/bpb.b15-01013.
PM, Biesiadecki BJ, Hummel JD, Weiss R, Fedorov VV. Adenosine-
578. Chen YJ, Chen SA, Chang MS, Lin CI. Arrhythmogenic activity of
induced atrial fibrillation: localized reentrant drivers in lateral right
cardiac muscle in pulmonary veins of the dog: implication for the
atria due to heterogeneous expression of adenosine a1 receptors
genesis of atrial fibrillation. Cardiovasc Res 48: 265–273, 2000.
and GIRK4 subunits in the human heart. Circulation 134: 486–498,
doi:10.1016/s0008-6363(00)00179-6.
2016. doi:10.1161/CIRCULATIONAHA.115.021165.
579. Ehrlich JR, Cha TJ, Zhang L, Chartier D, Villeneuve L, He bert TE,
565. Workman AJ, Kane KA, Rankin AC. Ionic basis of a differential effect Nattel S. Characterization of a hyperpolarization-activated time-de-
of adenosine on refractoriness in rabbit AV nodal and atrial isolated pendent potassium current in canine cardiomyocytes from pulmo-
myocytes. Cardiovasc Res 43: 974–984, 1999. doi:10.1016/s0008- nary vein myocardial sleeves and left atrium. J Physiol 557: 583–
6363(99)00166-2. 597, 2004. doi:10.1113/jphysiol.2004.061119.
566. Pau D, Workman AJ, Kane KA, Rankin AC. Electrophysiological and 580. Chen YC, Chen SA, Chen YJ, Chang MS, Chan P, Lin CI. Effects of
arrhythmogenic effects of 5-hydroxytryptamine on human atrial thyroid hormone on the arrhythmogenic activity of pulmonary vein
cells are reduced in atrial fibrillation. J Mol Cell Cardiol 42: 54–62, cardiomyocytes. J Am Coll Cardiol 39: 366–372, 2002. doi:10.
2007. doi:10.1016/j.yjmcc.2006.08.007. 1016/S0735-1097(01)01731-4.

1156 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

581. Tsuneoka Y, Irie M, Tanaka Y, Sugimoto T, Kobayashi Y, Kusakabe Medicine, edited by Zipes DP, Libby P, Bonow RO. Amsterdam:
T, Kato K, Hamaguchi S, Namekata I, Tanaka H. Permissive role of Elsevier, 2019, p. 619–647.
reduced inwardly-rectifying potassium current density in the auto-
maticity of the guinea pig pulmonary vein myocardium. J 595. Yuill KH, Hancox JC. Characteristics of single cells isolated from
Pharmacol Sci 133: 195–202, 2017. doi:10.1016/j.jphs.2016.12.006. the atrioventricular node of the adult guinea-pig heart. Pflugers
Arch 445: 311–320, 2002. doi:10.1007/s00424-002-0932-8.
582. Okamoto Y, Kawamura K, Nakamura Y, Ono K. Pathological impact
of hyperpolarization-activated chloride current peculiar to rat pul- 596. Hancox JC, Levi AJ, Lee CO, Heap P. A method for isolating rabbit
monary vein cardiomyocytes. J Mol Cell Cardiol 66: 53–62, 2014. atrioventricular node myocytes which retain normal morphology
doi:10.1016/j.yjmcc.2013.11.002. and function. Am J Physiol Heart Circ Physiol 265: H755–H766,
1993. doi:10.1152/ajpheart.1993.265.2.H755.
583. Chan CS, Chen YC, Chang SL, Lin YK, Kao YH, Chen SA, Chen YJ.
Heart failure differentially modulates the effects of ivabradine on 597. Hancox JC, Patel KC, Jones JV. Antiarrhythmics—from cell to clinic:
the electrical activity of the sinoatrial node and pulmonary veins. J past, present, and future. Heart 84: 14–24, 2000. doi:10.1136/heart.
Card Fail 24: 763–772, 2018. doi:10.1016/j.cardfail.2018.09.016. 84.1.14.

584. Chang CJ, Cheng CC, Yang TF, Chen YC, Lin YK, Chen SA, Chen 598. Meijler FL, Janse MJ. Morphology and electrophysiology of the
YJ. Selective and non-selective non-steroidal anti-inflammatory mammalian atrioventricular node. Physiol Rev 68: 608–647, 1988.
drugs differentially regulate pulmonary vein and atrial arrhythmo- doi:10.1152/physrev.1988.68.2.608.
genesis. Int J Cardiol 184: 559–567, 2015. doi:10.1016/j.ijcard.2015.
599. Munk AA, Adjemian RA, Zhao J, Ogbaghebriel A, Shrier A.
03.066.
Electrophysiological properties of morphologically distinct cells iso-
585. Lu YY, Lin YK, Wen ZH, Chen YC, Chen SA, Chen YJ. Latrunculin B lated from the rabbit atrioventricular node. J Physiol 493: 801–818,
modulates electrophysiological characteristics and arrhythmogen- 1996. doi:10.1113/jphysiol.1996.sp021424.
esis in pulmonary vein cardiomyocytes. Clin Sci (Lond) 130: 721–
732, 2016. doi:10.1042/CS20150593. 600. Atkinson AJ, Logantha SJ, Hao G, Yanni J, Fedorenko O, Sinha A,
Gilbert SH, Benson AP, Buckley DL, Anderson RH, Boyett MR,
586. Sicouri S, Antzelevitch C. Pulmonary vein sleeves as a pharmaco- Dobrzynski H. Functional, anatomical, and molecular investigation
logic model for the study of atrial fibrillation. Electrofisiol Arritm 3: of the cardiac conduction system and arrhythmogenic atrioventric-
108–113, 2010. ular ring tissue in the rat heart. J Am Heart Assoc 2: e000246,
2013. doi:10.1161/JAHA.113.000246.
587. Sicouri S, Cordeiro JM, Talarico M, Antzelevitch C. Antiarrhythmic
effects of losartan and enalapril in canine pulmonary vein sleeve 601. Greener ID, Tellez JO, Dobrzynski H, Yamamoto M, Graham GM,
preparations. J Cardiovasc Electrophysiol 22: 698–705, 2011. doi: Billeter R, Boyett MR. Ion channel transcript expression at the rabbit
10.1111/j.1540-8167.2010.01972.x. atrioventricular conduction axis. Circ Arrhythm Electrophysiol 2:
305–315, 2009. doi:10.1161/CIRCEP.108.803569.
588. Chan CS, Lin YK, Kao YH, Chen YC, Chen SA, Chen YJ. Hydrogen
sulphide increases pulmonary veins and atrial arrhythmogenesis 602. Hancox JC, Mitcheson JS. Ion channel and exchange currents in
with activation of protein kinase C. J Cell Mol Med 22: 3503–3513, single myocytes isolated from the rabbit atrioventricular node. Can
2018. doi:10.1111/jcmm.13627. J Cardiol 13: 1175–1182, 1997.

589. Lin YK, Lai MS, Chen YC, Cheng CC, Huang JH, Chen SA, Chen YJ, 603. Markowitz SM, Lerman BB. A contemporary view of atrioventricular
Lin CI. Hypoxia and reoxygenation modulate the arrhythmogenic nodal physiology. J Interv Card Electrophysiol 52: 271–279, 2018.
activity of the pulmonary vein and atrium. Clin Sci (Lond) 122: 121– doi:10.1007/s10840-018-0392-5.
132, 2012. doi:10.1042/CS20110178.
604. Belardinelli L, Shryock JC, Song Y, Wang D, Srinivas M. Ionic basis
590. Chang CJ, Cheng CC, Chen YC, Higa S, Huang JH, Chen SA, Chen of the electrophysiological actions of adenosine on cardiomyo-
YJ. Factor Xa inhibitors differently modulate electrical activities in cytes. FASEB J 9: 359–365, 1995. doi:10.1096/fasebj.9.5.7896004.
pulmonary veins and the sinoatrial node. Eur J Pharmacol 833:
462–471, 2018. doi:10.1016/j.ejphar.2018.07.003. 605. Cheng H, Curtis AE, Fellingham C, Hancox JC. Multiple ion channel
block by the cation channel inhibitor SKF-96365 in myocytes from
591. Lin WS, Tai CT, Hsieh MH, Tsai CF, Lin YK, Tsao HM, Huang JL, Yu the rabbit atrioventricular node. Physiol Rep 4: e12819, 2016.
WC, Yang SP, Ding YA, Chang MS, Chen SA. Catheter ablation of doi:10.14814/phy2.12819.
paroxysmal atrial fibrillation initiated by non-pulmonary vein ectopy.
Circulation 107: 3176–3183, 2003. doi:10.1161/01.CIR.0000074206. 606. Martynyuk AE, Kane KA, Cobbe SM, Rankin AC. Adenosine
52056.2D. increases potassium conductance in isolated rabbit atrioventricular
nodal myocytes. Cardiovasc Res 30: 668–675, 1995. doi:10.1016/
592. Miyazaki S, Kuwahara T, Kobori A, Takahashi Y, Takei A, Sato A, S0008-6363(95)00078-X.
Isobe M, Takahashi A. Long-term clinical outcome of extensive pul-
monary vein isolation-based catheter ablation therapy in patients 607. Orito K, Satoh K, Taira N. Cardiovascular profile of Ro 40-5967, a
with paroxysmal and persistent atrial fibrillation. Heart 97: 668– new nondihydropyridine calcium antagonist, delineated in isolated,
673, 2011. doi:10.1136/hrt.2009.186874. blood-perfused dog hearts. J Cardiovasc Pharmacol 22: 293–299,
1993. doi:10.1097/00005344-199308000-00019.
593. Liu J, Noble PJ, Xiao G, Abdelrahman M, Dobrzynski H, Boyett MR,
Lei M, Noble D. Role of pacemaking current in cardiac nodes: 608. Wang D, Shryock JC, Belardinelli L. Cellular basis for the negative
insights from a comparative study of sinoatrial node and atrioven- dromotropic effect of adenosine on rabbit single atrioventricular
tricular node. Prog Biophys Mol Biol 96: 294–304, 2008. doi:10. nodal cells. Circ Res 78: 697–706, 1996. doi:10.1161/01.RES.78.4.
1016/j.pbiomolbio.2007.07.009. 697.

594. Tomaselli GF, Rubart M, Zipes D. Mechanisms of cardiac arrhyth- 609. Zhang Y. In vivo recording of Zhang’s phenomenon (His electro-
mias. In: Braunwald’s Heart Disease A Textbook of Cardiovascular gram alternans): a novel index of atrioventricular node dual

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1157


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

pathway conduction. J Interv Card Electrophysiol 40: 99–103, than in ventricles. Cardiovasc Res 65: 117–127, 2005. doi:10.1016/j.
2014. doi:10.1007/s10840-014-9905-z. cardiores.2004.08.017.

610. Janse MJ, van Capelle FJ, Freud GE, Durrer D. Circus movement 626. Jalife J, Sicouri S, Delmar M, Michaels DC. Electrical uncoupling
within the AV node as a basis for supraventricular tachycardia as and impulse propagation in isolated sheep Purkinje fibers. Am J
shown by multiple microelectrode recording in the isolated rabbit Physiol Heart Circ Physiol 257: H179–H189, 1989. doi:10.1152/
heart. Circ Res 28: 403–414, 1971. doi:10.1161/01.res.28.4.403. ajpheart.1989.257.1.H179.

611. Freilich A, Tepper D. Adenosine and its cardiovascular effects. Am 627. Sheets MF, Hanck DA, Fozzard HA. Nonlinear relation between
Heart J 123: 1324–1328, 1992. doi:10.1016/0002-8703(92)91040-8. Vmax and INa in canine cardiac Purkinje cells. Circ Res 63: 386–
398, 1988. doi:10.1161/01.res.63.2.386.
612. Martynyuk AE, Seubert CN, Zima A, Morey TE, Belardinelli L, Lin G,
Cucchiara RF, Dennis DM. Contribution of IK,ADO to the negative 628. Spach MS, Miller WT 3rd, Miller-Jones E, Warren RB, Barr RC.
dromotropic effect of adenosine. Basic Res Cardiol 97: 286–294, Extracellular potentials related to intracellular action potentials dur-
2002. doi:10.1007/s00395-002-0349-8. ing impulse conduction in anisotropic canine cardiac muscle. Circ
Res 45: 188–204, 1979. doi:10.1161/01.res.45.2.188.
613. Cheng H, Li J, James AF, Inada S, Choisy SC, Orchard CH, Zhang
H, Boyett MR, Hancox JC. Characterization and influence of cardiac 629. Kanter HL, Laing JG, Beau SL, Beyer EC, Saffitz JE. Distinct patterns
background sodium current in the atrioventricular node. J Mol Cell of connexin expression in canine Purkinje fibers and ventricular
Cardiol 97: 114–124, 2016. doi:10.1016/j.yjmcc.2016.04.014. muscle. Circ Res 72: 1124–1131, 1993. doi:10.1161/01.RES.72.5.1124.

614. Hancox J, Levi A. The hyperpolarisation-activated current, If, is 630. Han W, Bao W, Wang Z, Nattel S. Comparison of ion-channel subunit
not required for pacemaking in single cells from the rabbit atrio- expression in canine cardiac Purkinje fibers and ventricular muscle.
ventricular node. Pflugers Arch 427: 121–128, 1994. doi:10.1007/ Circ Res 91: 790–797, 2002. doi:10.1161/01.res.0000039534.18114.d9.
BF00585950. 631. Szigeti G, Rusznák Z, Kovács L, Papp Z. Calcium-activated transient
615. Boyden PA. Purkinje physiology and pathophysiology. J Interv membrane currents are carried mainly by chloride ions in isolated
Card Electrophysiol 52: 255–262, 2018. doi:10.1007/s10840-018- atrial, ventricular and Purkinje cells of rabbit heart. Exp Physiol 83:
137–153, 1998. doi:10.1113/expphysiol.1998.sp004097.
0414-3.
632. Verkerk AO, Veldkamp MW, Bouman LN, van Ginneken AC.
616. Dun W, Boyden PA. The Purkinje cell; 2008 style. J Mol Cell
Calcium-activated Cl- current contributes to delayed afterdepolari-
Cardiol 45: 617–624, 2008. doi:10.1016/j.yjmcc.2008.08.001.
zations in single Purkinje and ventricular myocytes. Circulation 101:
617. Adamantidis MM, Caron JF, Dupuis BA. Triggered activity induced 2639–2644, 2000. doi:10.1161/01.cir.101.22.2639.
by combined mild hypoxia and acidosis in guinea-pig Purkinje
633. Rosati B, Dun W, Hirose M, Boyden PA, McKinnon D. Molecular ba-
fibers. J Mol Cell Cardiol 18: 1287–1299, 1986. doi:10.1016/s0022-
sis of the T- and L-type Ca21 currents in canine Purkinje fibres. J
2828(86)80432-1.
Physiol 579: 465–471, 2007. doi:10.1113/jphysiol.2006.127480.
618. Friedman PL, Stewart JR, Fenoglio JJ Jr, Wit AL. Survival of suben-
634. DiFrancesco D. Characterization of the pace-maker current kinetics
docardial Purkinje fibers after extensive myocardial infarction in
in calf Purkinje fibres. J Physiol 348: 341–367, 1984. doi:10.1113/
dogs. Circ Res 33: 597–611, 1973. doi:10.1161/01.res.33.5.597.
jphysiol.1984.sp015114.
619. Boyden PA, Barbhaiya C, Lee T, ter Keurs HE. Nonuniform Ca21
635. Cordeiro JM, Spitzer KW, Giles WR. Repolarizing K1 currents in rab-
transients in arrhythmogenic Purkinje cells that survive in the bit heart Purkinje cells. J Physiol 508: 811–823, 1998. doi:10.1111/
infarcted canine heart. Cardiovasc Res 57: 681–693, 2003. j.1469-7793.1998.811bp.x.
doi:10.1016/S0008-6363(02)00725-3.
636. Dumaine R, Cordeiro JM. Comparison of K1 currents in cardiac
620. Rosen MR, Gelband H, Hoffman BF. Effects of phentolamine on Purkinje cells isolated from rabbit and dog. J Mol Cell Cardiol 42:
electrophysiologic properties of isolated canine purkinje fibers. J 378–389, 2007. doi:10.1016/j.yjmcc.2006.10.019.
Pharmacol Exp Ther 179: 586–593, 1971.
637. Gadsby DC, Wit AL, Cranefield PF. The effects of acetylcholine on
621. Colatsky TJ. Voltage clamp measurements of sodium channel the electrical activity of canine cardiac Purkinje fibers. Circ Res 43:
properties in rabbit cardiac Purkinje fibres. J Physiol 305: 215–234, 29–35, 1978. doi:10.1161/01.res.43.1.29.
1980. doi:10.1113/jphysiol.1980.sp013359.
638. Callewaert G, Carmeliet E, Vereecke J. Single cardiac Purkinje
622. Papp Z, Sipido KR, Callewaert G, Carmeliet E. Two components of cells: general electrophysiology and voltage-clamp analysis of the
[Ca21]i-activated Cl- current during large [Ca21]i transients in single pace-maker current. J Physiol 349: 643–661, 1984. doi:10.1113/
rabbit heart Purkinje cells. J Physiol 483: 319–330, 1995. jphysiol.1984.sp015179.
doi:10.1113/jphysiol.1995.sp020588.
639. DiFrancesco D, Noble D. A model of cardiac electrical activity incor-
623. McGillivray RM, Wald RW. Measurement of the maximum rate of porating ionic pumps and concentration changes. Philos Trans R
rise of the cardiac action potential Vmax. Med Biol Eng Comput Soc Lond B Biol Sci 307: 353–398, 1985. doi:10.1098/rstb.1985.
22: 275–276, 1984. doi:10.1007/BF02442757. 0001.

624. Haufe V, Chamberland C, Dumaine R. The promiscuous nature of 640. Vassalle M, Yu H, Cohen IS. The pacemaker current in cardiac
the cardiac sodium current. J Mol Cell Cardiol 42: 469–477, 2007. Purkinje myocytes. J Gen Physiol 106: 559–578, 1995. doi:10.1085/
doi:10.1016/j.yjmcc.2006.12.005. jgp.106.3.559.

625. Haufe V, Cordeiro JM, Zimmer T, Wu YS, Schiccitano S, Benndorf 641. Boyden PA, Pu J, Pinto J, Keurs HE. Ca21 transients and Ca21
K, Dumaine R. Contribution of neuronal sodium channels to the car- waves in purkinje cells: role in action potential initiation. Circ Res
diac fast sodium current INa is greater in dog heart Purkinje fibers 86: 448–455, 2000. doi:10.1161/01.res.86.4.448.

1158 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

642. Tranum-Jensen J, Wilde AA, Vermeulen JT, Janse MJ. Morphology in distribution of ion channels in canine and human ventricular myo-
of electrophysiologically identified junctions between Purkinje cardium. Cardiovasc Res 65: 851–860, 2005. doi:10.1016/j.
fibers and ventricular muscle in rabbit and pig hearts. Circ Res 69: cardiores.2004.11.022.
429–437, 1991. doi:10.1161/01.res.69.2.429.
656. Antzelevitch C. Modulation of transmural repolarization. Ann NY
643. Nattel S, Quantz MA. Pharmacological response of quinidine Acad Sci 1047: 314–323, 2005. doi:10.1196/annals.1341.028.
induced early afterdepolarisations in canine cardiac Purkinje fibres:
insights into underlying ionic mechanisms. Cardiovasc Res 22: 657. Litovsky SH, Antzelevitch C. Transient outward current prominent
808–817, 1988. doi:10.1093/cvr/22.11.808. in canine ventricular epicardium but not endocardium. Circ Res 62:
116–126, 1988. doi:10.1161/01.res.62.1.116.
644. Janse MJ, Kleber AG, Capucci A, Coronel R, Wilms-Schopman F.
Electrophysiological basis for arrhythmias caused by acute ische- 658. Zygmunt AC, Eddlestone GT, Thomas GP, Nesterenko VV,
mia. Role of the subendocardium. J Mol Cell Cardiol 18: 339–355, Antzelevitch C. Larger late sodium conductance in M cells contrib-
1986. doi:10.1016/s0022-2828(86)80898-7. utes to electrical heterogeneity in canine ventricle. Am J Physiol
Heart Circ Physiol 281: H689–H697, 2001. doi:10.1152/ajpheart.
645. Cordeiro JM, Zeina T, Goodrow R, Kaplan AD, Thomas LM, 2001.281.2.H689.
Nesterenko VV, Treat JA, Hawel L 3rd, Byus C, Bett GC,
Rasmusson RL, Panama BK. Regional variation of the inwardly recti- 659. Zygmunt AC, Goodrow RJ, Antzelevitch C. INaCa contributes to elec-
fying potassium current in the canine heart and the contributions to trical heterogeneity within the canine ventricle. Am J Physiol Heart
differences in action potential repolarization. J Mol Cell Cardiol 84: Circ Physiol 278: H1671–H1678, 2000. doi:10.1152/ajpheart.2000.
52–60, 2015. doi:10.1016/j.yjmcc.2015.04.010. 278.5.H1671.

646. Liu DW, Gintant GA, Antzelevitch C. Ionic bases for electrophysio- 660. Liu DW, Antzelevitch C. Characteristics of the delayed rectifier cur-
logical distinctions among epicardial, midmyocardial, and endocar- rent (IKr and IKs) in canine ventricular epicardial, midmyocardial,
dial myocytes from the free wall of the canine left ventricle. Circ and endocardial myocytes. A weaker IKs contributes to the longer
Res 72: 671–687, 1993. doi:10.1161/01.res.72.3.671. action potential of the M cell. Circ Res 76: 351–365, 1995.
doi:10.1161/01.res.76.3.351.
647. Verduyn SC, Vos MA, van der Zande J, Kulcsàr A, Wellens HJ.
Further observations to elucidate the role of interventricular disper- 661. Anyukhovsky EP, Sosunov EA, Rosen MR. Regional differences in
sion of repolarization and early afterdepolarizations in the genesis electrophysiological properties of epicardium, midmyocardium,
of acquired torsade de pointes arrhythmias: a comparison between and endocardium. In vitro and in vivo correlations. Circulation 94:
almokalant and d-sotalol using the dog as its own control. J 1981–1988, 1996. doi:10.1161/01.CIR.94.8.1981.
Am Coll Cardiol 30: 1575–1584, 1997. doi:10.1016/S0735-1097(97)
00333-1. 662. Boukens BJ, Meijborg VM, Belterman CN, Opthof T, Janse MJ,
Schuessler RB, Coronel R, Efimov IR. Local transmural action poten-
€tjens RL, Wellens HJ, Vos
648. Volders PG, Sipido KR, Carmeliet E, Spa tial gradients are absent in the isolated, intact dog heart but pres-
MA. Repolarizing K1 currents ITO1 and IKs are larger in right than ent in the corresponding coronary-perfused wedge. Physiol Rep 5:
left canine ventricular midmyocardium. Circulation 99: 206–210, e13251, 2017. doi:10.14814/phy2.13251.
1999. doi:10.1161/01.cir.99.2.206.
663. Di Diego JM, Sicouri S, Myles RC, Burton FL, Smith GL, Antzelevitch
649. Di Diego JM, Sun ZQ, Antzelevitch C. Ito and action potential notch C. Optical and electrical recordings from isolated coronary-per-
are smaller in left vs. right canine ventricular epicardium. Am J fused ventricular wedge preparations. J Mol Cell Cardiol 54: 53–
Physiol Heart Circ Physiol 271: H548–H561, 1996. doi:10.1152/ 64, 2013. doi:10.1016/j.yjmcc.2012.10.017.
ajpheart.1996.271.2.H548.
664. Yan GX, Shimizu W, Antzelevitch C. Characteristics and distribution
650. Calloe K, Aistrup GL, Di Diego JM, Goodrow RJ, Treat JA, Cordeiro of M cells in arterially perfused canine left ventricular wedge prepa-
JM. Interventricular differences in sodium current and its potential rations. Circulation 98: 1921–1927, 1998. doi:10.1161/01.cir.98.18.
role in Brugada syndrome. Physiol Rep 6: e13787, 2018. doi:10. 1921.
14814/phy2.13787.
665. Janse MJ, Coronel R, Opthof T, Sosunov EA, Anyukhovsky EP,
651. Osadchii OE. Arrhythmogenic drugs can amplify spatial heteroge-
Rosen MR. Repolarization gradients in the intact heart: transmural
neities in the electrical restitution in perfused guinea-pig heart: an
or apico-basal? Prog Biophys Mol Biol 109: 6–15, 2012. doi:10.1016/
evidence from assessments of monophasic action potential dura-
j.pbiomolbio.2012.03.001.
tions and JT intervals. PLoS One 13: e0191514, 2018. doi:10.1371/
journal.pone.0191514. 666. Meijborg VM, Conrath CE, Opthof T, Belterman CN, de Bakker JM,
Coronel R. Electrocardiographic T wave and its relation with ven-
652. Bueno-Orovio A, Hanson BM, Gill JS, Taggart P, Rodriguez B. In
tricular repolarization along major anatomical axes. Circ Arrhythm
vivo human left-to-right ventricular differences in rate adaptation
Electrophysiol 7: 524–531, 2014. doi:10.1161/CIRCEP.113.001622.
transiently increase pro-arrhythmic risk following rate acceleration.
PLoS One 7: e52234, 2012. doi:10.1371/journal.pone.0052234. 667. Anderson RD, Kumar S, Parameswaran R, Wong G, Voskoboinik A,
Sugumar H, Watts T, Sparks PB, Morton JB, McLellan A, Kistler PM,
653. Sekiya S, Ichikawa S, Tsutsumi T, Harumi K. Nonuniform action
Kalman J, Lee G. Differentiating right- and left-sided outflow tract
potential durations at different sites in canine left ventricle. Jpn
ventricular arrhythmias: classical ECG signatures and prediction
Heart J 24: 935–945, 1983. doi:10.1536/ihj.24.935.
algorithms. Circ Arrhythm Electrophysiol 12: e007392, 2019.
654. Sicouri S, Antzelevitch C. A subpopulation of cells with unique elec- doi:10.1161/CIRCEP.119.007392.
trophysiological properties in the deep subepicardium of the ca-
nine ventricle. The M cell. Circ Res 68: 1729–1741, 1991. doi:10.1161/ 668. Ashino S, Watanabe I, Kofune M, Nagashima K, Ohkubo K,
01.res.68.6.1729. Okumura Y, Nakai T, Kasamaki Y, Hirayama A. Abnormal action
potential duration restitution property in the right ventricular out-
655. Szentadrassy N, Banyasz T, Biro T, Szabo G, Toth BI, Magyar J, flow tract in Brugada syndrome. Circ J 74: 664–670, 2010.
Lazar J, Varro A, Kovacs L, Nanasi PP. Apico-basal inhomogeneity doi:10.1253/circj.CJ-09-0872.

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1159


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

669. Lu YY, Cheng CC, Tsai CF, Lin YK, Lee TI, Chen YC, Chen SA, Chen in heart failure patients with and without atrial fibrillation. Front
YJ. Discrepant effects of heart failure on electrophysiological prop- Physiol 9: 1383, 2018. doi:10.3389/fphys.2018.01383.
erty in right ventricular outflow tract and left ventricular outflow tract
cardiomyocytes. Clin Sci (Lond) 131: 1317–1327, 2017. doi:10.1042/ 682. Carmeliet E. Repolarisation and frequency in cardiac cells. J
CS20170121. Physiol (Paris) 73: 903–923, 1977.

670. Morita H, Zipes DP, Morita ST, Wu J. Differences in arrhythmogenic- 683. Li GR, Du XL, Siow YL, O K, Tse HF, Lau CP. Calcium-activated tran-
ity between the canine right ventricular outflow tract and anteroin- sient outward chloride current and phase 1 repolarization of swine
ferior right ventricle in a model of Brugada syndrome. Heart ventricular action potential. Cardiovasc Res 58: 89–98, 2003.
Rhythm 4: 66–74, 2007. doi:10.1016/j.hrthm.2006.08.030. doi:10.1016/s0008-6363(02)00859-3.

671. Lu YY, Chung FP, Chen YC, Tsai CF, Kao YH, Chao TF, Huang JH, 684. Schultz JH, Volk T, Bassaláy P, Hennings JC, H€
ubner CA, Ehmke H.
Chen SA, Chen YJ. Distinctive electrophysiological characteristics Molecular and functional characterization of Kv4.2 and KChIP2
of right ventricular out-flow tract cardiomyocytes. J Cell Mol Med expressed in the porcine left ventricle. Pflugers Arch 454: 195–
18: 1540–1548, 2014. doi:10.1111/jcmm.12329. 207, 2007. doi:10.1007/s00424-006-0203-1.

672. Huang CL. Murine electrophysiological models of cardiac arrhyth- 685. Jost N, Virág L, Comtois P, Ördo € g B, Szuts V, Sepre
 nyi G, Bitay M,
mogenesis. Physiol Rev 97: 283–409, 2017. doi:10.1152/physrev. Kohajda Z, Koncz I, Nagy N, Sze l T, Magyar J, Kovács M, Puskás
00007.2016. LG, Lengyel C, Wettwer E, Ravens U, Nánási PP, Papp JG, Varro  A,
Nattel S. Ionic mechanisms limiting cardiac repolarization reserve
673. Boukens BJ, Rivaud MR, Rentschler S, Coronel R. Misinterpretation in humans compared to dogs. J Physiol 591: 4189–4206, 2013.
of the mouse ECG: ‘musing the waves of Mus musculus’. J Physiol doi:10.1113/jphysiol.2013.261198.
592: 4613–4626, 2014. doi:10.1113/jphysiol.2014.279380.
686. Kopljar I, Lu HR, Van Ammel K, Otava M, Tekle F, Teisman A,
674. Nerbonne JM, Nichols CG, Schwarz TL, Escande D. Genetic manip- Gallacher DJ. Development of a human iPSC cardiomyocyte-based
ulation of cardiac K1 channel function in mice: what have we scoring system for cardiac hazard identification in early drug safety
learned, and where do we go from here? Circ Res 89: 944–956, de-risking. Stem Cell Rep 11: 1365–1377, 2018. doi:10.1016/j.stemcr.
2001. doi:10.1161/hh2301.100349. 2018.11.007.

675. Wang L, Feng ZP, Kondo CS, Sheldon RS, Duff HJ. Developmental 687. Mummery CL. Perspectives on the use of human induced pluripo-
changes in the delayed rectifier K1 channels in mouse heart. Circ tent stem cell-derived cardiomyocytes in biomedical research.
Res 79: 79–85, 1996. doi:10.1161/01.RES.79.1.79. Stem Cell Rep 11: 1306–1311, 2018. doi:10.1016/j.stemcr.2018.11.011.
676. Brunner M, Peng X, Liu GX, Ren XQ, Ziv O, Choi BR, Mathur R, 688. Brodehl A, Ebbinghaus H, Deutsch MA, Gummert J, Gartner A,
Hajjiri M, Odening KE, Steinberg E, Folco EJ, Pringa E, Centracchio Ratnavadivel S, Milting H. Human induced pluripotent stem-cell-
J, Macharzina RR, Donahay T, Schofield L, Rana N, Kirk M, Mitchell derived cardiomyocytes as models for genetic cardiomyopathies.
GF, Poppas A, Zehender M, Koren G. Mechanisms of cardiac Int J Mol Sci 20: 4381, 2019. doi:10.3390/ijms20184381.
arrhythmias and sudden death in transgenic rabbits with long QT
syndrome. J Clin Invest 118: 2246–2259, 2008. doi:10.1172/ 689. Casini S, Verkerk AO, Remme CA. Human iPSC-derived cardiomyo-
JCI33578. cytes for investigation of disease mechanisms and therapeutic
strategies in inherited arrhythmia syndromes: strengths and limita-
677. Major P, Baczko  I, Hiripi L, Odening KE, Juhász V, Kohajda Z, tions. Cardiovasc Drugs Ther 31: 325–344, 2017. doi:10.1007/
Horváth A, Sepre nyi G, Kovács M, Virág L, Jost N, Prorok J, Ördo €g s10557-017-6735-0.
B, Doleschall Z, Nattel S, Varro A, Bo
 sze Z. A novel transgenic rab-
bit model with reduced repolarization reserve: long QT syndrome 690. Garg P, Garg V, Shrestha R, Sanguinetti MC, Kamp TJ, Wu JC.
caused by a dominant-negative mutation of the KCNE1 gene. Br J Human induced pluripotent stem cell-derived cardiomyocytes as
Pharmacol 173: 2046–2061, 2016. doi:10.1111/bph.13500. models for cardiac channelopathies: a primer for non-electrophysi-
ologists. Circ Res 123: 224–243, 2018. doi:10.1161/CIRCRESAHA.
678. Odening KE, Bodi I, Franke G, Rieke R, Ryan de Medeiros A, Perez- 118.311209.
Feliz S, F€
urniss H, Mettke L, Michaelides K, Lang CN, Steinfurt J,
Pantulu ND, Ziupa D, Menza M, Zehender M, Bugger H, Peyronnet 691. Karakikes I, Ameen M, Termglinchan V, Wu JC. Human induced
R, Behrends JC, Doleschall Z, Zur Hausen A, Bode C, Jolivet G, pluripotent stem cell-derived cardiomyocytes: insights into molecu-
Brunner M. Transgenic short-QT syndrome 1 rabbits mimic the lar, cellular, and functional phenotypes. Circ Res 117: 80–88, 2015.
human disease phenotype with QT/action potential duration short- doi:10.1161/CIRCRESAHA.117.305365.
ening in the atria and ventricles and increased ventricular tachycar-
692. Horváth A, Lemoine MD, Lo € ser A, Mannhardt I, Flenner F, Uzun AU,
dia/ventricular fibrillation inducibility. Eur Heart J 40: 842–853,
2019. doi:10.1093/eurheartj/ehy761. Neuber C, Breckwoldt K, Hansen A, Girdauskas E, Reichenspurner
H, Willems S, Jost N, Wettwer E, Eschenhagen T, Christ T. Low rest-
679. Martinez-Mateu L, Saiz J, Aromolaran AS. Differential modulation of ing membrane potential and low inward rectifier potassium cur-
IK and ICa,L channels in high-fat diet-induced obese guinea pig rents are not inherent features of hiPSC-derived cardiomyocytes.
atria. Front Physiol 10: 1212, 2019. doi:10.3389/fphys.2019.01212. Stem Cell Rep 10: 822–833, 2018. doi:10.1016/j.stemcr.2018.01.012.

680. Pustovit KB, Potekhina VM, Ivanova AD, Petrov AM, Abramochkin 693. Horvath B, Banyasz T, Jian Z, Hegyi B, Kistamas K, Nanasi PP, Izu
DV, Kuzmin VS. Extracellular ATP and beta-NAD alter electrical LT, Chen-Izu Y. Dynamics of the late Na1 current during cardiac
properties and cholinergic effects in the rat heart in age-specific action potential and its contribution to afterdepolarizations. J Mol
manner. Purinergic Signal 15: 107–117, 2019. doi:10.1007/s11302- Cell Cardiol 64: 59–68, 2013. doi:10.1016/j.yjmcc.2013.08.010.
019-09645-6.
694. Verkerk AO, Remme CA. ‘Mature’ resting membrane potentials in
681. Molina CE, Abu-Taha IH, Wang Q, Rosello  -Díez E, Kamler M, Nattel human-induced pluripotent stem cell-derived cardiomyocytes: fact
S, Ravens U, Wehrens XH, Hove-Madsen L, Heijman J, Dobrev D. or artefact? Europace 21: 1928, 2019. doi:10.1093/europace/
Profibrotic, electrical, and calcium-handling remodeling of the atria euz243.

1160 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

695. Angsutararux P, Luanpitpong S, Chingsuwanrote P, Supraditaporn 708. Luo CH, Rudy Y. A dynamic model of the cardiac ventricular action
K, Waeteekul S, Terbto P, Lorthongpanich C, Laowtammathron C, potential. II. Afterdepolarizations, triggered activity, and potentia-
U-Pratya Y, Issaragrisil S. Generation of human induced pluripotent tion. Circ Res 74: 1097–1113, 1994. doi:10.1161/01.res.74.6.1097.
stem cell line carrying SCN5AC2204>T Brugada mutation
709. Tomek J, Rodriguez B, Bub G, Heijman J. beta-Adrenergic receptor
(MUSli009-A-1) introduced by CRISPR/Cas9-mediated genome
stimulation inhibits proarrhythmic alternans in postinfarction border
editing. Stem Cell Res 41: 101618, 2019. doi:10.1016/j.scr.2019.
zone cardiomyocytes: a computational analysis. Am J Physiol
101618.
Heart Circ Physiol 313: H338–H353, 2017. doi:10.1152/ajpheart.
696. Ben Jehuda R, Shemer Y, Binah O. Genome editing in induced plu- 00094.2017.
ripotent stem cells using CRISPR/Cas9. Stem Cell Rev Rep 14:
710. Trayanova NA. Whole-heart modeling: applications to cardiac elec-
323–336, 2018. doi:10.1007/s12015-018-9811-3.
trophysiology and electromechanics. Circ Res 108: 113–128, 2011.
697. Mesquita FC, Arantes PC, Kasai-Brunswick TH, Araujo DS, Gubert doi:10.1161/CIRCRESAHA.110.223610.
F, Monnerat G, Silva Dos Santos D, Neiman G, Leitão IC, Barbosa 711. Vandersickel N, Van Nieuwenhuyse E, Seemann G, Panfilov AV.
RA, Coutinho JL, Vaz IM, Dos Santos MN, Borgonovo T, Cruz FE, Spatial patterns of excitation at tissue and whole organ level due to
Miriuka S, Medei EH, Campos de Carvalho AC, Carvalho AB. early afterdepolarizations. Front Physiol 8: 404, 2017. doi:10.3389/
R534C mutation in hERG causes a trafficking defect in iPSC- fphys.2017.00404.
derived cardiomyocytes from patients with type 2 long QT syn-
drome. Sci Rep 9: 19203, 2019. doi:10.1038/s41598-019-55837-w. 712. Weiss JN, Karma A, Shiferaw Y, Chen PS, Garfinkel A, Qu Z. From
pulsus to pulseless: the saga of cardiac alternans. Circ Res 98:
 F, Boni M, Crotti
698. Mura M, Bastaroli F, Corli M, Ginevrino M, Calabro 1244–1253, 2006. doi:10.1161/01.RES.0000224540.97431.f0.
L, Valente EM, Schwartz PJ, Gnecchi M. Generation of the human
induced pluripotent stem cell (hiPSC) line PSMi006-A from a patient 713. Dutta S, Chang KC, Beattie KA, Sheng J, Tran PN, Wu WW, Wu M,
affected by an autosomal recessive form of long QT syndrome Strauss DG, Colatsky T, Li Z. Optimization of an in silico cardiac cell
type 1. Stem Cell Res 42: 101658, 2020. doi:10.1016/j.scr.2019. model for proarrhythmia risk assessment. Front Physiol 8: 616,
101658. 2017. doi:10.3389/fphys.2017.00616.

€ ser A, Lemoine MD, Christ T, Scherschel K, Meyer C,


699. Krause J, Lo 714. Li Z, Dutta S, Sheng J, Tran PN, Wu W, Chang K, Mdluli T, Strauss
Blankenberg S, Zeller T, Eschenhagen T, Stenzig J. Rat atrial engi- DG, Colatsky T. Improving the in silico assessment of proarrhythmia
neered heart tissue: a new in vitro model to study atrial biology. risk by combining hERG (human Ether-a-go-go-Related gene) chan-
Basic Res Cardiol 113: 41, 2018. doi:10.1007/s00395-018-0701-2. nel-drug binding kinetics and multichannel pharmacology. Circ
Arrhythm Electrophysiol 10: e004628, 2017. doi:10.1161/CIRCEP.
700. Park SJ, Zhang D, Qi Y, Li Y, Lee KY, Bezzerides VJ, Yang P, Xia S, 116.004628.
Kim SL, Liu X, Lu F, Pasqualini FS, Campbell PH, Geva J, Roberts
715. Passini E, Britton OJ, Lu HR, Rohrbacher J, Hermans AN, Gallacher
AE, Kleber AG, Abrams DJ, Pu WT, Parker KK. Insights into the
DJ, Greig RJ, Bueno-Orovio A, Rodriguez B. Human in silico drug
pathogenesis of catecholaminergic polymorphic ventricular tachy-
trials demonstrate higher accuracy than animal models in predict-
cardia from engineered human heart tissue. Circulation 140: 390–
ing clinical pro-arrhythmic cardiotoxicity. Front Physiol 8: 668,
404, 2019. doi:10.1161/CIRCULATIONAHA.119.039711.
2017. doi:10.3389/fphys.2017.00668.
701. Pr€
uller J, Mannhardt I, Eschenhagen T, Zammit PS, Figeac N.
716. Carusi A, Burrage K, Rodríguez B. Bridging experiments, models
Satellite cells delivered in their niche efficiently generate functional
and simulations: an integrative approach to validation in computa-
myotubes in three-dimensional cell culture. PLoS One 13: tional cardiac electrophysiology. Am J Physiol Heart Circ Physiol
e0202574, 2018. doi:10.1371/journal.pone.0202574. 303: H144–H155, 2012. doi:10.1152/ajpheart.01151.2011.
702. Noble D. Successes and failures in modeling heart cell electrophys- 717. Tomek J, Bueno-Orovio A, Passini E, Zhou X, Minchole A, Britton O,
iology. Heart Rhythm 8: 1798–1803, 2011. doi:10.1016/j.hrthm.2011. Bartolucci C, Severi S, Shrier A, Virag L, Varro A, Rodriguez B.
06.014. Development, calibration, and validation of a novel human ventricu-
lar myocyte model in health, disease, and drug block. Elife 8:
703. Noble D. A modification of the Hodgkin–Huxley equations applica-
e48890, 2019. doi:10.7554/eLife.48890.
ble to Purkinje fibre action and pace-maker potentials. J Physiol
160: 317–352, 1962. doi:10.1113/jphysiol.1962.sp006849. 718. Beattie KA, Hill AP, Bardenet R, Cui Y, Vandenberg JI, Gavaghan
DJ, de Boer TP, Mirams GR. Sinusoidal voltage protocols for rapid
704. Beeler GW, Reuter H. Reconstruction of the action potential of ven-
characterisation of ion channel kinetics. J Physiol 596: 1813–1828,
tricular myocardial fibres. J Physiol 268: 177–210, 1977. doi:10.1113/ 2018. doi:10.1113/JP275733.
jphysiol.1977.sp011853.
719. Lu Y, Mahaut-Smith MP, Varghese A, Huang CL, Kemp PR,
705. Hodgkin AL, Huxley AF. A quantitative description of membrane Vandenberg JI. Effects of premature stimulation on HERG K1 chan-
current and its application to conduction and excitation in nerve. J nels. J Physiol 537: 843–851, 2001. doi:10.1111/j.1469-7793.2001.
Physiol 117: 500–544, 1952. doi:10.1113/jphysiol.1952.sp004764. 00843.x.
706. Dutta S, Minchole A, Zacur E, Quinn TA, Taggart P, Rodriguez B. 720. Clancy CE, Tateyama M, Kass RS. Insights into the molecular mech-
Early afterdepolarizations promote transmural reentry in ischemic anisms of bradycardia-triggered arrhythmias in long QT-3 syn-
human ventricles with reduced repolarization reserve. Prog drome. J Clin Invest 110: 1251–1262, 2002. doi:10.1172/JCI15928.
Biophys Mol Biol 120: 236–248, 2016. doi:10.1016/j.pbiomolbio.
2016.01.008. 721. Mahajan A, Shiferaw Y, Sato D, Baher A, Olcese R, Xie LH, Yang
MJ, Chen PS, Restrepo JG, Karma A, Garfinkel A, Qu Z, Weiss JN. A
707. Luo CH, Rudy Y. A dynamic model of the cardiac ventricular action rabbit ventricular action potential model replicating cardiac dynam-
potential. I. Simulations of ionic currents and concentration ics at rapid heart rates. Biophys J 94: 392–410, 2008. doi:10.1529/
changes. Circ Res 74: 1071–1096, 1994. doi:10.1161/01.res.74.6.1071. biophysj.106.98160.

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1161


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

722. Ramasubramanian S, Rudy Y. The structural basis of IKs ion-chan- update from a Cardiac Safety Research Consortium/Health and
nel activation: mechanistic insights from molecular simulations. Environmental Sciences Institute/FDA meeting. Ther Innov Regul
Biophys J 114: 2584–2594, 2018. doi:10.1016/j.bpj.2018.04.023. Sci 53: 519–525, 2019. doi:10.1177/2168479018795117.

723. Xu J, Rudy Y. Effects of beta-subunit on gating of a potassium ion 738. Courtemanche M, Ramirez RJ, Nattel S. Ionic mechanisms underly-
channel: molecular simulations of cardiac IKs activation. J Mol Cell ing human atrial action potential properties: insights from a mathe-
Cardiol 124: 35–44, 2018. doi:10.1016/j.yjmcc.2018.10.003. matical model. Am J Physiol Heart Circ Physiol 275: H301–H321,
1998. doi:10.1152/ajpheart.1998.275.1.H301.
724. Hund TJ, Rudy Y. Rate dependence and regulation of action poten-
tial and calcium transient in a canine cardiac ventricular cell model. 739. Nygren A, Fiset C, Firek L, Clark JW, Lindblad DS, Clark RB, Giles
Circulation 110: 3168–3174, 2004. doi:10.1161/01.CIR.0000147231. WR. Mathematical model of an adult human atrial cell: the role of
69595.D3. K1 currents in repolarization. Circ Res 82: 63–81, 1998. doi:10.1161/
01.res.82.1.63.
725. Zeng J, Laurita KR, Rosenbaum DS, Rudy Y. Two components of
the delayed rectifier K1 current in ventricular myocytes of the 740. Lindblad DS, Murphey CR, Clark JW, Giles WR. A model of the
guinea pig type. Theoretical formulation and their role in repolariza- action potential and underlying membrane currents in a rabbit atrial
tion. Circ Res 77: 140–152, 1995. doi:10.1161/01.res.77.1.140. cell. Am Physiol Heart Circ J Physiol 271: H1666–H1696, 1996.
doi:10.1152/ajpheart.1996.271.4.H1666.
726. Decker KF, Heijman J, Silva JR, Hund TJ, Rudy Y. Properties and
ionic mechanisms of action potential adaptation, restitution, and 741. Grandi E, Pandit SV, Voigt N, Workman AJ, Dobrev D, Jalife J, Bers
accommodation in canine epicardium. Am J Physiol Heart Circ DM. Human atrial action potential and Ca21 model: sinus rhythm
Physiol 296: H1017–H1026, 2009. doi:10.1152/ajpheart.01216.2008. and chronic atrial fibrillation. Circ Res 109: 1055–1066, 2011.
doi:10.1161/CIRCRESAHA.111.253955.
727. Decker KF, Rudy Y. Ionic mechanisms of electrophysiological het-
erogeneity and conduction block in the infarct border zone. Am J 742. Koivuma€ki JT, Korhonen T, Tavi P. Impact of sarcoplasmic reticulum
Physiol Heart Circ Physiol 299: H1588–H1597, 2010. doi:10.1152/ calcium release on calcium dynamics and action potential morphol-
ajpheart.00362.2010. ogy in human atrial myocytes: a computational study. PLoS
Comput Biol 7: e1001067, 2011. doi:10.1371/journal.pcbi.1001067.
728. Heijman J, Volders PG, Westra RL, Rudy Y. Local control of beta-ad-
renergic stimulation: effects on ventricular myocyte electrophysiol- 743. Maleckar MM, Greenstein JL, Trayanova NA, Giles WR.
ogy and Ca21-transient. J Mol Cell Cardiol 50: 863–871, 2011. Mathematical simulations of ligand-gated and cell-type specific
doi:10.1016/j.yjmcc.2011.02.007. effects on the action potential of human atrium. Prog Biophys Mol
729. Shannon TR, Wang F, Puglisi J, Weber C, Bers DM. A mathematical Biol 98: 161–170, 2008. doi:10.1016/j.pbiomolbio.2009.01.010.
treatment of integrated Ca dynamics within the ventricular myo- 744. Grandi E, Maleckar MM. Anti-arrhythmic strategies for atrial fibrilla-
cyte. Biophys J 87: 3351–3371, 2004. doi:10.1529/ tion: the role of computational modeling in discovery, development,
biophysj.104.047449. and optimization. Pharmacol Ther 168: 126–142, 2016. doi:10.1016/j.
730. Bondarenko VE, Szigeti GP, Bett GC, Kim SJ, Rasmusson RL. pharmthera.2016.09.012.
Computer model of action potential of mouse ventricular myocytes. 745. Vagos M, van Herck IG, Sundnes J, Arevalo HJ, Edwards AG,
Am J Physiol Heart Circ Physiol 287: H1378–H1403, 2004. €aki JT. Computational modeling of electrophysiology and
Koivuma
doi:10.1152/ajpheart.00185.2003.
pharmacotherapy of atrial fibrillation: recent advances and future
731. Morotti S, Edwards AG, McCulloch AD, Bers DM, Grandi E. A novel challenges. Front Physiol 9: 1221, 2018. doi:10.3389/fphys.2018.
computational model of mouse myocyte electrophysiology to 01221.
assess the synergy between Na1 loading and CaMKII. J Physiol
746. Wilhelms M, Hettmann H, Maleckar MM, Koivuma €ki JT, Do
€ ssel O,
592: 1181–1197, 2014. doi:10.1113/jphysiol.2013.266676.
Seemann G. Benchmarking electrophysiological models of human
732. ten Tusscher KH, Noble D, Noble PJ, Panfilov AV. A model for atrial myocytes. Front Physiol 3: 487, 2012. doi:10.3389/fphys.
human ventricular tissue. Am J Physiol Heart Circ Physiol 286: 2012.00487.
H1573–H1589, 2004. doi:10.1152/ajpheart.00794.2003.
747. Stewart P, Aslanidi OV, Noble D, Noble PJ, Boyett MR, Zhang H.
733. ten Tusscher KH, Panfilov AV. Alternans and spiral breakup in a Mathematical models of the electrical action potential of Purkinje
human ventricular tissue model. Am J Physiol Heart Circ Physiol fibre cells. Philos Trans A Math Phys Eng Sci 367: 2225–2255,
291: H1088–H1100, 2006. doi:10.1152/ajpheart.00109.2006. 2009. doi:10.1098/rsta.2008.0283.

734. Grandi E, Pasqualini FS, Bers DM. A novel computational model of 748. Li J, Logantha SJ, Yanni J, Cai X, Zhang H, Dobrzynski H, Hart G,
the human ventricular action potential and Ca transient. J Mol Cell Boyett M. Computer simulation of Purkinje fibres from a rabbit
Cardiol 48: 112–121, 2010. doi:10.1016/j.yjmcc.2009.09.019. model of congestive heart failure. Computing in Cardiology 2013.
Zaragoza, Spain, 2013, p. 361–364.
735. Carro J, Rodríguez JF, Laguna P, Pueyo E. A human ventricular cell
model for investigation of cardiac arrhythmias under hyperkalaemic 749. Corrias A, Giles W, Rodriguez B. Ionic mechanisms of electrophysi-
conditions. Philos Trans A Math Phys Eng Sci 369: 4205–4232, ological properties and repolarization abnormalities in rabbit
2011. doi:10.1098/rsta.2011.0127. Purkinje fibers. Am J Physiol Heart Circ Physiol 300: H1806–
H1813, 2011. doi:10.1152/ajpheart.01170.2010.
736. O’Hara T, Virág L, Varro  A, Rudy Y. Simulation of the undiseased
human cardiac ventricular action potential: model formulation and 750. Behradfar E, Nygren A, Vigmond EJ. The role of Purkinje-myocar-
experimental validation. PLoS Comput Biol 7: e1002061, 2011. dial coupling during ventricular arrhythmia: a modeling study. PLoS
doi:10.1371/journal.pcbi.1002061. One 9: e88000, 2014. doi:10.1371/journal.pone.0088000.

737. Strauss DG, Gintant G, Li Z, Wu W, Blinova K, Vicente J, Turner JR, 751. Sampson KJ, Iyer V, Marks AR, Kass RS. A computational model of
Sager PT. Comprehensive in vitro proarrhythmia assay (CiPA) Purkinje fibre single cell electrophysiology: implications for the

1162 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

long QT syndrome. J Physiol 588: 2643–2655, 2010. doi:10.1113/ ischaemic ventricular tachycardia. Interface Focus 1: 396–407,
jphysiol.2010.187328. 2011. doi:10.1098/rsfs.2010.0041.

752. Trovato C, Passini E, Nagy N, Varro  A, Abi-Gerges N, Severi S, 766. Bueno-Orovio A, Kay D, Grau V, Rodriguez B, Burrage K. Fractional
Rodriguez B. Human Purkinje in silico model enables mechanistic diffusion models of cardiac electrical propagation: role of structural
investigations into automaticity and pro-arrhythmic abnormalities. J heterogeneity in dispersion of repolarization. J R Soc Interface 11:
Mol Cell Cardiol 142: 24–38, 2020. doi:10.1016/j.yjmcc.2020.04. 20140352, 2014. doi:10.1098/rsif.2014.0352.
001.
767. Patel RB, Ng J, Reddy V, Chokshi M, Parikh K, Subacius H,
753. Severi S, Fantini M, Charawi LA, DiFrancesco D. An updated com- Alsheikh-Ali AA, Nguyen T, Link MS, Goldberger JJ, Ilkhanoff L,
putational model of rabbit sinoatrial action potential to investigate Kadish AH. Early repolarization associated with ventricular arrhyth-
the mechanisms of heart rate modulation. J Physiol 590: 4483– mias in patients with chronic coronary artery disease. Circ
4499, 2012. doi:10.1113/jphysiol.2012.229435. Arrhythm Electrophysiol 3: 489–495, 2010. doi:10.1161/CIRCEP.
109.921130.
754. Zhang H, Holden AV, Kodama I, Honjo H, Lei M, Varghese T,
Boyett MR. Mathematical models of action potentials in the periph- 768. Britton OJ, Bueno-Orovio A, Van Ammel K, Lu HR, Towart R,
ery and center of the rabbit sinoatrial node. Am J Physiol Heart Gallacher DJ, Rodriguez B. Experimentally calibrated population of
Circ Physiol 279: H397–H421, 2000. doi:10.1152/ajpheart.2000. models predicts and explains intersubject variability in cardiac cel-
279.1.H397. lular electrophysiology. Proc Natl Acad Sci USA 110: E2098–
E2105, 2013. doi:10.1073/pnas.1304382110.
755. Fabbri A, Fantini M, Wilders R, Severi S. Computational analysis of
the human sinus node action potential: model development and 769. Groenendaal W, Ortega FA, Kherlopian AR, Zygmunt AC, Krogh-
effects of mutations. J Physiol 595: 2365–2396, 2017. doi:10.1113/ Madsen T, Christini DJ. Cell-specific cardiac electrophysiology
JP273259. models. PLoS Comput Biol 11: e1004242, 2015. doi:10.1371/journal.
pcbi.1004242.
756. Yaniv Y, Lakatta EG, Maltsev VA. From two competing oscillators to
one coupled-clock pacemaker cell system. Front Physiol 6: 28, 770. Johnstone RH, Chang ET, Bardenet R, de Boer TP, Gavaghan DJ,
2015. doi:10.3389/fphys.2015.00028. Pathmanathan P, Clayton RH, Mirams GR. Uncertainty and variabili-
ty in models of the cardiac action potential: can we build trustwor-
757. Paci M, Hyttinen J, Aalto-Seta €la
€ K, Severi S. Computational models thy models? J Mol Cell Cardiol 96: 49–62, 2016. doi:10.1016/j.
of ventricular- and atrial-like human induced pluripotent stem cell yjmcc.2015.11.018.
derived cardiomyocytes. Ann Biomed Eng 41: 2334–2348, 2013.
doi:10.1007/s10439-013-0833-3. 771. Ni H, Morotti S, Grandi E. A heart for diversity: simulating variability
in cardiac arrhythmia research. Front Physiol 9: 958, 2018.
758. Koivuma €ki JT, Naumenko N, Tuomainen T, Takalo J, Oksanen M, doi:10.3389/fphys.2018.00958.
Puttonen KA, Lehtonen S, Kuusisto J, Laakso M, Koistinaho J, Tavi
P. Structural immaturity of human iPSC-derived cardiomyocytes: in 772. Pueyo E, Dangerfield CE, Britton OJ, Virág L, Kistamás K,
silico investigation of effects on function and disease modeling. Szentandrássy N, Jost N, Varro  A, Nánási PP, Burrage K, Rodríguez
Front Physiol 9: 80, 2018. doi:10.3389/fphys.2018.00080. B. Experimentally-based computational investigation into beat-to-
beat variability in ventricular repolarization and its response to ionic
€ lo
759. Paci M, Po € nen RP, Cori D, Penttinen K, Aalto-Seta
€la
€ K, Severi S, current inhibition. PLoS One 11: e0151461, 2016. doi:10.1371/journal.
Hyttinen J. Automatic optimization of an in silico model of human pone.0151461.
iPSC derived cardiomyocytes recapitulating calcium handling
abnormalities. Front Physiol 9: 709, 2018. doi:10.3389/fphys.2018. 773. Sarkar AX, Christini DJ, Sobie EA. Exploiting mathematical models
00709. to illuminate electrophysiological variability between individuals. J
Physiol 590: 2555–2567, 2012. doi:10.1113/jphysiol.2011.223313.
760. Colman MA, Pinali C, Trafford AW, Zhang H, Kitmitto A. A computa-
774. Muszkiewicz A, Britton OJ, Gemmell P, Passini E, Sánchez C, Zhou
tional model of spatio-temporal cardiac intracellular calcium han-
X, Carusi A, Quinn TA, Burrage K, Bueno-Orovio A, Rodriguez B.
dling with realistic structure and spatial flux distribution from
Variability in cardiac electrophysiology: using experimentally-cali-
sarcoplasmic reticulum and t-tubule reconstructions. PLoS Comput
brated populations of models to move beyond the single virtual
Biol 13: e1005714, 2017. doi:10.1371/journal.pcbi.1005714.
physiological human paradigm. Prog Biophys Mol Biol 120: 115–
761. Restrepo JG, Karma A. Spatiotemporal intracellular calcium dynam- 127, 2016. doi:10.1016/j.pbiomolbio.2015.12.002.
ics during cardiac alternans. Chaos 19: 037115, 2009. doi:10.1063/
775. Muszkiewicz A, Liu X, Bueno-Orovio A, Lawson BA, Burrage K,
1.3207835.
Casadei B, Rodriguez B. From ionic to cellular variability in human
762. Sato D, Bers DM, Shiferaw Y. Formation of spatially discordant atrial myocytes: an integrative computational and experimental
alternans due to fluctuations and diffusion of calcium. PLoS One 8: study. Am J Physiol Heart Circ Physiol 314: H895–H916, 2018.
e85365, 2013. doi:10.1371/journal.pone.0085365. doi:10.1152/ajpheart.00477.2017.

763. Fenton F, Karma A. Vortex dynamics in three-dimensional continu- 776. Passini E, Minchole A, Coppini R, Cerbai E, Rodriguez B, Severi S,
ous myocardium with fiber rotation: filament instability and fibrilla- Bueno-Orovio A. Mechanisms of pro-arrhythmic abnormalities in
tion. Chaos 8: 20–47, 1998. doi:10.1063/1.166311. ventricular repolarisation and anti-arrhythmic therapies in human
hypertrophic cardiomyopathy. J Mol Cell Cardiol 96: 72–81, 2016.
764. Bueno-Orovio A, Cherry EM, Fenton FH. Minimal model for human doi:10.1016/j.yjmcc.2015.09.003.
ventricular action potentials in tissue. J Theor Biol 253: 544–560,
2008. doi:10.1016/j.jtbi.2008.03.029. 777. Reilly SN, Liu X, Carnicer R, Recalde A, Muszkiewicz A, Jayaram R,
Carena MC, Wijesurendra R, Stefanini M, Surdo NC, Lomas O,
765. Relan J, Chinchapatnam P, Sermesant M, Rhode K, Ginks M, Ratnatunga C, Sayeed R, Krasopoulos G, Rajakumar T, Bueno-
Delingette H, Rinaldi CA, Razavi R, Ayache N. Coupled personaliza- Orovio A, Verheule S, Fulga TA, Rodriguez B, Schotten U, Casadei
tion of cardiac electrophysiology models for prediction of B. Up-regulation of miR-31 in human atrial fibrillation begets the

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1163


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

arrhythmia by depleting dystrophin and neuronal nitric oxide syn- 792. Wei-Qing H, Qing-Nuan K, Lin X, Cheng-Hao G, Qi-Yi Z. Expression
thase. Sci Transl Med 8: 340ra, 2016. doi:10.1126/scitranslmed. of hyperpolarization-activated cyclic nucleotide-gated cation chan-
aac4296. nel (HCN4) is increased in hypertrophic cardiomyopathy.
Cardiovasc Pathol 20: 110–113, 2011. doi:10.1016/j.carpath.2010.
778. Brugada J, Campuzano O, Arbelo E, Sarquella-Brugada G, Brugada 01.007.
R. present Status of Brugada syndrome: JACC State-of-the-Art
Review. J Am Coll Cardiol 72: 1046–1059, 2018. doi:10.1016/j. 793. Marban E, Robinson SW, Wier WG. Mechanisms of arrhythmogenic
jacc.2018.06.037. delayed and early afterdepolarizations in ferret ventricular muscle.
J Clin Invest 78: 1185–1192, 1986. doi:10.1172/JCI112701.
779. Li KH, Lee S, Yin C, Liu T, Ngarmukos T, Conte G, Yan GX, Sy RW,
Letsas KP, Tse G. Brugada syndrome: a comprehensive review of 794. Vandersickel N, de Boer TP, Vos MA, Panfilov AV. Perpetuation of
pathophysiological mechanisms and risk stratification strategies. torsade de pointes in heterogeneous hearts: competing foci or re-
Int J Cardiol Heart Vasc 26: 100468, 2020. doi:10.1016/j.ijcha. entry? J Physiol 594: 6865–6878, 2016. doi:10.1113/JP271728.
2020.100468.
795. Morotti S, Grandi E, Summa A, Ginsburg KS, Bers DM. Theoretical
780. Bueno-Orovio A, Cherry EM, Evans SJ, Fenton FH. Basis for the study of L-type Ca21 current inactivation kinetics during action
induction of tissue-level phase-2 reentry as a repolarization disor- potential repolarization and early afterdepolarizations. J Physiol
der in the Brugada syndrome. Biomed Res Int 2015: 197586, 2015. 590: 4465–4481, 2012. doi:10.1113/jphysiol.2012.231886.
doi:10.1155/2015/197586. 796. Weiss JN, Garfinkel A, Karagueuzian HS, Chen PS, Qu Z. Early
781. Cascio WE, Johnson TA, Gettes LS. Electrophysiologic changes in afterdepolarizations and cardiac arrhythmias. Heart Rhythm 7:
ischemic ventricular myocardium: I. Influence of ionic, metabolic, 1891–1899, 2010. doi:10.1016/j.hrthm.2010.09.017.
and energetic changes. J Cardiovasc Electrophysiol 6: 1039– 797. Kurata Y, Tsumoto K, Hayashi K, Hisatome I, Tanida M, Kuda Y,
1062, 1995. doi:10.1111/j.1540-8167.1995.tb00381.x. Shibamoto T. Dynamical mechanisms of phase-2 early afterdepola-
rizations in human ventricular myocytes: insights from bifurcation
782. Janse MJ, D’Alnoncourt CN. Reflections on reentry and focal activ-
analyses of two mathematical models. Am J Physiol Heart Circ
ity. Am J Cardiol 60: 21F–26F, 1987. doi:10.1016/0002-9149(87)
Physiol 312: H106–H127, 2017. doi:10.1152/ajpheart.00115.2016.
90716 -8.
798. Clancy CE, Tateyama M, Liu H, Wehrens XH, Kass RS. Non-equilib-
783. Kleber AG, Riegger CB, Janse MJ. Extracellular K1 and H1 shifts in
rium gating in cardiac Na1 channels: an original mechanism of ar-
early ischemia: mechanisms and relation to changes in impulse
rhythmia. Circulation 107: 2233–2237, 2003. doi:10.1161/01.CIR.
propagation. J Mol Cell Cardiol 19: 35–44, 1987. doi:10.1016/
0000069273.51375.BD.
S0022-2828(87)80608-9.
799. Dai L, Zang Y, Zheng D, Xia L, Gong Y. Role of CaMKII and PKA in
784. Pinto JM, Boyden PA. Electrical remodeling in ischemia and infarc- early afterdepolarization of human ventricular myocardium cell: a
tion. Cardiovasc Res 42: 284–297, 1999. doi:10.1016/s0008-6363 computational model study. Comput Math Methods Med 2016:
(99)00013-9. 4576313, 2016. doi:10.1155/2016/4576313.
785. Wit AL, Hoffman BF, Cranefield PF. Slow conduction, reentry, and 800. Uchinoumi H, Yang Y, Oda T, Li N, Alsina KM, Puglisi JL, Chen-Izu
the mechanism of ventricular arrhythmias in myocardial infarction. Y, Cornea RL, Wehrens XH, Bers DM. CaMKII-dependent phospho-
Bull NY Acad Med 47: 1233–1234, 1971. rylation of RyR2 promotes targetable pathological RyR2 conforma-
tional shift. J Mol Cell Cardiol 98: 62–72, 2016. doi:10.1016/j.
786. Ruskin JN. The cardiac arrhythmia suppression trial (CAST). N Engl
yjmcc.2016.06.007.
J Med 321: 386–388, 1989. doi:10.1056/NEJM198908103210608.
801. van Oort RJ, McCauley MD, Dixit SS, Pereira L, Yang Y, Respress
787. Singh BN. Controlling cardiac arrhythmias: to delay conduction or
JL, Wang Q, De Almeida AC, Skapura DG, Anderson ME, Bers DM,
to prolong refractoriness. Cardiovasc Drugs Ther 3: 671–674,
Wehrens XH. Ryanodine receptor phosphorylation by calcium/cal-
1989. doi:10.1007/BF01857618.
modulin-dependent protein kinase II promotes life-threatening ven-
788. Cerbai E, Pino R, Porciatti F, Sani G, Toscano M, Maccherini M, tricular arrhythmias in mice with heart failure. Circulation 122:
Giunti G, Mugelli A. Characterization of the hyperpolarization-acti- 2669–2679, 2010. doi:10.1161/CIRCULATIONAHA.110.982298.
vated current, If, in ventricular myocytes from human failing heart. 802. Luo M, Anderson ME. Mechanisms of altered Ca21 handling in heart
Circulation 95: 568–571, 1997. doi:10.1161/01.cir.95.3.568. failure. Circ Res 113: 690–708, 2013. doi:10.1161/CIRCRESAHA.113.
301651.
789. Stillitano F, Lonardo G, Zicha S, Varro A, Cerbai E, Mugelli A, Nattel
S. Molecular basis of funny current (If) in normal and failing human 803. Voigt N, Li N, Wang Q, Wang W, Trafford AW, Abu-Taha I, Sun Q,
heart. J Mol Cell Cardiol 45: 289–299, 2008. doi:10.1016/j.yjmcc. Wieland T, Ravens U, Nattel S, Wehrens XH, Dobrev D. Enhanced
2008.04.013. sarcoplasmic reticulum Ca21 leak and increased Na1-Ca21
exchanger function underlie delayed afterdepolarizations in
790. Stillitano F, Lonardo G, Giunti G, Del Lungo M, Coppini R, Spinelli V,
patients with chronic atrial fibrillation. Circulation 125: 2059–2070,
Sartiani L, Poggesi C, Mugelli A, Cerbai E. Chronic atrial fibrillation
2012. doi:10.1161/CIRCULATIONAHA.111.067306.
alters the functional properties of If in the human atrium. J
Cardiovasc Electrophysiol 24: 1391–1400, 2013. doi:10.1111/ 804. Gomis-Tena J, Saiz J. Role of Ca21-dependent Cl- current on
jce.12212. delayed afterdepolarizations. A simulation study. Ann Biomed Eng
36: 752–761, 2008. doi:10.1007/s10439-008-9460-9.
791. Riesen SC, Schober KE, Bonagura JD, Carnes CA. Myocardial
expression of hyperpolarization-activated, cyclic nucleotide-gated 805. Liu MB, Ko CY, Song Z, Garfinkel A, Weiss JN, Qu Z. A dynamical
proteins in healthy cats and cats with hypertrophic cardiomyopathy. threshold for cardiac delayed afterdepolarization-mediated trig-
Schweiz Arch Tierheilkd 155: 143–147, 2013. doi:10.1024/0036- gered activity. Biophys J 111: 2523–2533, 2016. doi:10.1016/j.
7281/a000431. bpj.2016.10.009.

1164 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

806. Chang SH, Chen YC, Chiang SJ, Higa S, Cheng CC, Chen YJ, Chen 821. Roston TM, Haji-Ghassemi O, LaPage MJ, Batra AS, Bar-Cohen Y,
SA. Increased Ca21 sparks and sarcoplasmic reticulum Ca21 stores Anderson C, Lau YR, Maginot K, Gebauer RA, Etheridge SP, Potts
potentially determine the spontaneous activity of pulmonary vein JE, Van Petegem F, Sanatani S. Catecholaminergic polymorphic
cardiomyocytes. Life Sci 83: 284–292, 2008. doi:10.1016/j.lfs. ventricular tachycardia patients with multiple genetic variants in the
2008.06.014. PACES CPVT Registry. PLoS One 13: e0205925, 2018. doi:10.1371/
journal.pone.0205925.
807. Cheng H, Lederer WJ. Calcium sparks. Physiol Rev 88: 1491–1545,
2008. doi:10.1152/physrev.00030.2007. 822. Johnson DM, Heijman J, Bode EF, Greensmith DJ, van der Linde H,
Abi-Gerges N, Eisner DA, Trafford AW, Volders PG. Diastolic spon-
808. Belevych AE, Ho HT, Bonilla IM, Terentyeva R, Schober KE, taneous calcium release from the sarcoplasmic reticulum increases
Terentyev D, Carnes CA, Gyo € rke S. The role of spatial organization beat-to-beat variability of repolarization in canine ventricular myo-
of Ca21 release sites in the generation of arrhythmogenic diastolic cytes after beta-adrenergic stimulation. Circ Res 112: 246–256,
Ca21 release in myocytes from failing hearts. Basic Res Cardiol 2013. doi:10.1161/CIRCRESAHA.112.275735.
112: 44, 2017. doi:10.1007/s00395-017-0633-2.
823. Marks AR, Priori S, Memmi M, Kontula K, Laitinen PJ. Involvement
809. Cheng H, Lederer MR, Lederer WJ, Cannell MB. Calcium sparks of the cardiac ryanodine receptor/calcium release channel in cate-
and [Ca21]i waves in cardiac myocytes. Am J Physiol Cell Physiol cholaminergic polymorphic ventricular tachycardia. J Cell Physiol
270: C148–C159, 1996. doi:10.1152/ajpcell.1996.270.1.C148. 190: 1–6, 2002. doi:10.1002/jcp.10031.

810. Niggli E, Lederer WJ. Voltage-independent calcium release in heart 824. Landstrom AP, Dobrev D, Wehrens XH. Calcium signaling and car-
muscle. Science 250: 565–568, 1990. doi:10.1126/science.2173135. diac arrhythmias. Circ Res 120: 1969–1993, 2017. doi:10.1161/
CIRCRESAHA.117.310083.
811. Baader AP, B€uchler L, Bircher-Lehmann L, Kleber AG. Real time,
825. Bányász T, Horváth B, Virág L, Bárándi L, Szentadrássy N, Harmati
confocal imaging of Ca21 waves in arterially perfused rat hearts.
G, Magyar J, Marangoni S, Zaza A, Varro  A, Nánási P. Reverse rate
Cardiovasc Res 53: 105–115, 2002. doi:10.1016/s0008-6363(01)
dependency is an intrinsic property of canine cardiac preparations.
00423-0.
Cardiovasc Res 84: 237–244, 2009. doi:10.1093/cvr/cvp213.
812. Nivala M, de Lange E, Rovetti R, Qu Z. Computational modeling 826. Boyett MR, Jewell BR. A study of the factors responsible for rate-
and numerical methods for spatiotemporal calcium cycling in ven- dependent shortening of the action potential in mammalian ventric-
tricular myocytes. Front Physiol 3: 114, 2012. doi:10.3389/ ular muscle. J Physiol 285: 359–380, 1978. doi:10.1113/jphysiol.
fphys.2012.00114. 1978.sp012576.
813. Nivala M, Ko CY, Nivala M, Weiss JN, Qu Z. The emergence of sub- 827. Elharrar V, Surawicz B. Cycle length effect on restitution of action
cellular pacemaker sites for calcium waves and oscillations. J potential duration in dog cardiac fibers. Am J Physiol Heart Circ
Physiol 591: 5305–5320, 2013. doi:10.1113/jphysiol.2013.259960. Physiol 244: H782–H792, 1983. doi:10.1152/ajpheart.1983.244.6.
H782.
814. Song Z, Ko CY, Nivala M, Weiss JN, Qu Z. Calcium-voltage coupling
in the genesis of early and delayed afterdepolarizations in cardiac 828. Ito S, Surawicz B. Effect of tetraethylammonium chloride on action
myocytes. Biophys J 108: 1908–1921, 2015. doi:10.1016/j.bpj.2015. potential in cardiac Purkinje fibers. Am J Physiol Heart Circ
03.011. Physiol 241: H139–H144, 1981. doi:10.1152/ajpheart.1981.241.2.H139.

ber AG, Riegger CB, Janse MJ. Electrical uncoupling and


815. Kle 829. Virág L, Acsai K, Hála O, Zaza A, Bitay M, Bogáts G, Papp JG, Varro 
increase of extracellular resistance after induction of ischemia in A. Self-augmentation of the lengthening of repolarization is related
isolated, arterially perfused rabbit papillary muscle. Circ Res 61: to the shape of the cardiac action potential: implications for reverse
271–279, 1987. doi:10.1161/01.res.61.2.271. rate dependency. Br J Pharmacol 156: 1076–1084, 2009.
doi:10.1111/j.1476-5381.2009.00116.x.
816. Xie Y, Sato D, Garfinkel A, Qu Z, Weiss JN. So little source, so much
sink: requirements for afterdepolarizations to propagate in tissue. 830. Zaza A. Control of the cardiac action potential: the role of repolari-
Biophys J 99: 1408–1415, 2010. doi:10.1016/j.bpj.2010.06.042. zation dynamics. J Mol Cell Cardiol 48: 106–111, 2010. doi:10.1016/j.
yjmcc.2009.07.027.
817. Rutherford SL, Trew ML, Sands GB, LeGrice IJ, Smaill BH. High-re-
831. Shattock MJ, Park KC, Yang HY, Lee AW, Niederer S, MacLeod KT,
solution 3-dimensional reconstruction of the infarct border zone:
Winter J. Restitution slope is principally determined by steady-state
impact of structural remodeling on electrical activation. Circ Res 111:
action potential duration. Cardiovasc Res 113: 817–828, 2017.
301–311, 2012. doi:10.1161/CIRCRESAHA.111.260943.
doi:10.1093/cvr/cvx063.
818. Wang Y, Joyner RW, Wagner MB, Cheng J, Lai D, Crawford BH.
832. Nash MP, Bradley CP, Sutton PM, Clayton RH, Kallis P, Hayward
Stretch-activated channel activation promotes early afterdepolari-
MP, Paterson DJ, Taggart P. Whole heart action potential duration
zations in rat ventricular myocytes under oxidative stress. Am J
restitution properties in cardiac patients: a combined clinical and
Physiol Heart Circ Physiol 296: H1227–H1235, 2009. doi:10.1152/ modelling study. Exp Physiol 91: 339–354, 2006. doi:10.1113/
ajpheart.00808.2008. expphysiol.2005.031070.
819. Ho€ fer T, Politi A, Heinrich R. Intercellular Ca21 wave propagation 833. Pak HN, Hong SJ, Hwang GS, Lee HS, Park SW, Ahn JC, Moo Ro Y,
through gap-junctional Ca21 diffusion: a theoretical study. Biophys Kim YH. Spatial dispersion of action potential duration restitution
J 80: 75–87, 2001. doi:10.1016/S0006-3495(01)75996-6. kinetics is associated with induction of ventricular tachycardia/fibril-
lation in humans. J Cardiovasc Electrophysiol 15: 1357–1363,
820. Bai J, Wang K, Liu Y, Li Y, Liang C, Luo G, Dong S, Yuan Y, Zhang H.
2004. doi:10.1046/j.1540-8167.2004.03569.x.
Computational cardiac modeling reveals mechanisms of ventricular
arrhythmogenesis in long QT syndrome type 8: CACNA1C R858H 834. Riccio ML, Koller ML, Gilmour RF Jr. Electrical restitution and spatio-
mutation linked to ventricular fibrillation. Front Physiol 8: 771, 2017. temporal organization during ventricular fibrillation. Circ Res 84:
doi:10.3389/fphys.2017.00771. 955–963, 1999. doi:10.1161/01.RES.84.8.955.

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1165


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

835. Robinson RB, Boyden PA, Hoffman BF, Hewett KW. Electrical resti- 850. Chudin E, Goldhaber J, Garfinkel A, Weiss J, Kogan B. Intracellular
tution process in dispersed canine cardiac Purkinje and ventricular Ca21 dynamics and the stability of ventricular tachycardia. Biophys
cells. Am J Physiol Heart Circ Physiol 253: H1018–H1025, 1987. J 77: 2930–2941, 1999. doi:10.1016/S0006-3495(99)77126-2.
doi:10.1152/ajpheart.1987.253.5.H1018.
851. Diáz ME, O’Neill SC, Eisner DA. Sarcoplasmic reticulum calcium
836. Weiss JN, Garfinkel A, Karagueuzian HS, Nguyen TP, Olcese R, content fluctuation is the key to cardiac alternans. Circ Res 94:
Chen PS Qu Z. Perspective: a dynamics-based classification of ven- 650–656, 2004. doi:10.1161/01.RES.0000119923.64774.72.
tricular arrhythmias. J Mol Cell Cardiol 82: 136–152, 2015. doi:10.
1016/j.yjmcc.2015.02.017. 852. Nolasco JB, Dahlen RW. A graphic method for the study of alterna-
tion in cardiac action potentials. J Appl Physiol 25: 191–196, 1968.
837. Garfinkel A, Kim YH, Voroshilovsky O, Qu Z, Kil JR, Lee MH, doi:10.1152/jappl.1968.25.2.191.
Karagueuzian HS, Weiss JN, Chen PS. Preventing ventricular fibril-
lation by flattening cardiac restitution. Proc Natl Acad Sci USA 97: 853. Pruvot EJ, Katra RP, Rosenbaum DS, Laurita KR. Role of calcium cy-
6061–6066, 2000. doi:10.1073/pnas.090492697. cling versus restitution in the mechanism of repolarization alter-
nans. Circ Res 94: 1083–1090, 2004. doi:10.1161/01.RES.00001
838. Osadchii OE. Role of abnormal repolarization in the mechanism of 25629.72053.95.
cardiac arrhythmia. Acta Physiol (Oxf) 220, Suppl 712: 1–71, 2017.
doi:10.1111/apha.12902. 854. Eisner DA, Choi HS, Diáz ME, O'Neill SC, Trafford AW. Integrative
analysis of calcium cycling in cardiac muscle. Circ Res 87: 1087–
839. Morgan JM, Cunningham D, Rowland E. Dispersion of monophasic 1094, 2000. doi:10.1161/01.res.87.12.1087.
action potential duration: demonstrable in humans after premature
ventricular extrastimulation but not in steady state. J Am Coll 855. Picht E, DeSantiago J, Blatter LA, Bers DM. Cardiac alternans do
Cardiol 19: 1244–1253, 1992. doi:10.1016/0735-1097(92)90331-g. not rely on diastolic sarcoplasmic reticulum calcium content fluctua-
tions. Circ Res 99: 740–748, 2006. doi:10.1161/01.RES.00002
840. Wu TJ, Lin SF, Weiss JN, Ting CT, Chen PS. Two types of ventricular 44002.88813.91.
fibrillation in isolated rabbit hearts: importance of excitability and
action potential duration restitution. Circulation 106: 1859–1866, 856. Qu Z, Liu MB, Nivala M. A unified theory of calcium alternans in ven-
2002. doi:10.1161/01.cir.0000031334.49170.fb. tricular myocytes. Sci Rep 6: 35625, 2016. doi:10.1038/srep35625.

841. Ni H, Zhang H, Grandi E, Narayan SM, Giles WR. Transient outward 857. Livshitz LM, Rudy Y. Regulation of Ca21 and electrical alternans in
K1 current can strongly modulate action potential duration and initi- cardiac myocytes: role of CAMKII and repolarizing currents. Am J
ate alternans in the human atrium. Am J Physiol Heart Circ Physiol Physiol Heart Circ Physiol 292: H2854–H2866, 2007. doi:10.1152/
316: H527–H542, 2019. doi:10.1152/ajpheart.00251.2018. ajpheart.01347.2006.
842. Tolkacheva EG, Anumonwo JM, Jalife J. Action potential duration 858. Tomek J, Tomková M, Zhou X, Bub G, Rodriguez B. Modulation of
restitution portraits of mammalian ventricular myocytes: role of cal- cardiac alternans by altered sarcoplasmic reticulum calcium
cium current. Biophys J 91: 2735–2745, 2006. doi:10.1529/ release: a simulation study. Front Physiol 9: 1306, 2018. doi:
biophysj.106.083865. 10.3389/fphys.2018.01306.
843. Kline RP, Morad M. Potassium efflux in heart muscle during activity: 859. Zhou X, Bueno-Orovio A, Orini M, Hanson B, Hayward M, Taggart
extracellular accumulation and its implications. J Physiol 280: 537– P, Lambiase PD, Burrage K, Rodriguez B. In vivo and in silico inves-
558, 1978. doi:10.1113/jphysiol.1978.sp012400. tigation into mechanisms of frequency dependence of repolariza-
844. Weiss JN, Nivala M, Garfinkel A, Qu Z. Alternans and arrhythmias: tion alternans in human ventricular cardiomyocytes. Circ Res 118:
from cell to heart. Circ Res 108: 98–112, 2011. doi:10.1161/ 266–278, 2016. doi:10.1161/CIRCRESAHA.115.307836.
CIRCRESAHA.110.223586. 860. Qu Z, Garfinkel A, Chen PS, Weiss JN. Mechanisms of discordant
845. Narayan SM, Franz MR, Clopton P, Pruvot EJ, Krummen DE. alternans and induction of reentry in simulated cardiac tissue.
Repolarization alternans reveals vulnerability to human atrial fibrillation. Circulation 102: 1664–1670, 2000. doi:10.1161/01.cir.102.14.1664.
Circulation 123: 2922–2930, 2011. doi:10.1161/CIRCULATIONAHA.
861. Thomsen MB, Truin M, van Opstal JM, Beekman JD, Volders PG,
110.977827.
Stengl M, Vos MA. Sudden cardiac death in dogs with remodeled
846. Wilson LD, Jeyaraj D, Wan X, Hoeker GS, Said TH, Gittinger M, hearts is associated with larger beat-to-beat variability of repolari-
Laurita KR, Rosenbaum DS. Heart failure enhances susceptibility to zation. Basic Res Cardiol 100: 279–287, 2005. doi:10.1007/
arrhythmogenic cardiac alternans. Heart Rhythm 6: 251–259, s00395-005-0519-6.
2009. doi:10.1016/j.hrthm.2008.11.008.
862. Ufret-Vincenty CA, Baro DJ, Lederer WJ, Rockman HA, Quinones
847. Tomek J, Hao G, Tomková M, Lewis A, Carr C, Paterson DJ, LE, Santana LF. Role of sodium channel deglycosylation in the gen-
Rodriguez B, Bub G, Herring N. beta-Adrenergic receptor stimula- esis of cardiac arrhythmias in heart failure. J Biol Chem 276:
tion and alternans in the border zone of a healed infarct: an ex vivo 28197–28203, 2001. doi:10.1074/jbc.M102548200.
study and computational investigation of arrhythmogenesis. Front
863. Shimoni Y, Emmett T, Schmidt R, Nygren A, Kargacin G. Sex-de-
Physiol 10: 350, 2019. doi:10.3389/fphys.2019.00350.
pendent impairment of cardiac action potential conduction in type 1
848. Wilson LD, Rosenbaum DS. Mechanisms of arrythmogenic cardiac diabetic rats. Am J Physiol Heart Circ Physiol 296: H1442–H1450,
alternans. Europace 9 Suppl 6: vi77–82, 2007. doi:10.1093/ 2009. doi:10.1152/ajpheart.01150.2008.
europace/eum210.
864. Yu P, Hu L, Xie J, Chen S, Huang L, Xu Z, Liu X, Zhou Q, Yuan P,
849. Pastore JM, Girouard SD, Laurita KR, Akar FG, Rosenbaum DS. Yan X, Jin J, Shen Y, Zhu W, Fu L, Chen Q, Yu J, Hu J, Cao Q, Wan
Mechanism linking T-wave alternans to the genesis of cardiac fibril- R, Hong K. O-GlcNAcylation of cardiac Nav1.5 contributes to the de-
lation. Circulation 99: 1385–1394, 1999. doi:10.1161/01.cir.99.10. velopment of arrhythmias in diabetic hearts. Int J Cardiol 260: 74–
1385. 81, 2018. doi:10.1016/j.ijcard.2018.02.099.

1166 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

865. Howarth FC, Qureshi MA, Hassan Z, Al Kury LT, Isaev D, Parekh K, 877. Ai X, Pogwizd SM. Connexin 43 downregulation and dephosphoryl-
Yammahi SR, Oz M, Adrian TE, Adeghate E. Changing pattern of ation in nonischemic heart failure is associated with enhanced
gene expression is associated with ventricular myocyte dysfunction colocalized protein phosphatase type 2A. Circ Res 96: 54–63,
and altered mechanisms of Ca21 signalling in young type 2 Zucker 2005. doi:10.1161/01.RES.0000152325.07495.5a.
diabetic fatty rat heart. Exp Physiol 96: 325–337, 2011. doi:10.1113/
878. Boulaksil M, Bierhuizen MF, Engelen MA, Stein M, Kok BJ, van
expphysiol.2010.055574.
Amersfoorth SC, Vos MA, van Rijen HV, de Bakker JM, van Veen
866. Zicha S, Maltsev VA, Nattel S, Sabbah HN, Undrovinas AI. Post-tran- TA. Spatial heterogeneity of Cx43 is an arrhythmogenic substrate
scriptional alterations in the expression of cardiac Na1 channel of polymorphic ventricular tachycardias during compensated car-
subunits in chronic heart failure. J Mol Cell Cardiol 37: 91–100, diac hypertrophy in rats. Front Cardiovasc Med 3: 5, 2016.
2004. doi:10.1016/j.yjmcc.2004.04.003. doi:10.3389/fcvm.2016.00005.

f A, Pacher P, Kocsis E, Koltay 879. Dupont E, Matsushita T, Kaba RA, Vozzi C, Coppen SR, Khan N,
867. Lengyel C, Virág L, Kovács PP, Kristo
Kaprielian R, Yacoub MH, Severs NJ. Altered connexin expression
ZM, Nánási PP, To  th M, Kecskeme  ti V, Papp JG, Varro
 A, Jost N.
in human congestive heart failure. J Mol Cell Cardiol 33: 359–371,
Role of slow delayed rectifier K1-current in QT prolongation in the
2001. doi:10.1006/jmcc.2000.1308.
alloxan-induced diabetic rabbit heart. Acta Physiol (Oxf) 192: 359–
368, 2008. doi:10.1111/j.1748-1716.2007.01753.x. 880. Allessie M, Ausma J, Schotten U. Electrical, contractile and struc-
tural remodeling during atrial fibrillation. Cardiovasc Res 54: 230–
868. Shimoni Y, Ewart HS, Severson D. Type I and II models of diabetes 246, 2002. doi:10.1016/s0008-6363(02)00258-4.
produce different modifications of K1 currents in rat heart: role of
insulin. J Physiol 507: 485–496, 1998. doi:10.1111/j.1469-7793.1998. 881. Ma ZG, Yuan YP, Wu HM, Zhang X, Tang QZ. Cardiac fibrosis: new
485bt.x. insights into the pathogenesis. Int J Biol Sci 14: 1645–1657, 2018.
doi:10.7150/ijbs.28103.
869. Zhang Y, Xiao J, Wang H, Luo X, Wang J, Villeneuve LR, Zhang H,
Bai Y, Yang B, Wang Z. Restoring depressed HERG K1 channel 882. Vaidya K, Semsarian C, Chan KH. Atrial fibrillation in hypertrophic
function as a mechanism for insulin treatment of abnormal QT pro- cardiomyopathy. Heart Lung Circ 26: 975–982, 2017. doi:10.1016/j.
longation and associated arrhythmias in diabetic rabbits. Am J hlc.2017.05.116.
Physiol Heart Circ Physiol 291: H1446–H1455, 2006. doi:10.1152/
883. Macia E, Dolmatova E, Cabo C, Sosinsky AZ, Dun W, Coromilas J,
ajpheart.01356.2005.
Ciaccio EJ, Boyden PA, Wit AL, Duffy HS. Characterization of gap
870. Van Wagoner DR, Pond AL, McCarthy PM, Trimmer JS, Nerbonne junction remodeling in epicardial border zone of healing canine
JM. Outward K1 current densities and Kv1.5 expression are infarcts and electrophysiological effects of partial reversal by roti-
gaptide. Circ Arrhythm Electrophysiol 4: 344–351, 2011. doi:10.
reduced in chronic human atrial fibrillation. Circ Res 80: 772–781,
1161/CIRCEP.110.959312.
1997. doi:10.1161/01.res.80.6.772.
884. Martins RP, Kaur K, Hwang E, Ramirez RJ, Willis BC, Filgueiras-
871. Heijman J, Kirchner D, Kunze F, Chre tien EM, Michel-Reher MB,
Rama D, Ennis SR, Takemoto Y, Ponce-Balbuena D, Zarzoso M,
Voigt N, Knaut M, Michel MC, Ravens U, Dobrev D. Muscarinic
O’Connell RP, Musa H, Guerrero-Serna G, Avula UM, Swartz MF,
type-1 receptors contribute to IK,ACh in human atrial cardiomyo- Bhushal S, Deo M, Pandit SV, Berenfeld O, Jalife J. Dominant fre-
cytes and are upregulated in patients with chronic atrial fibrillation. quency increase rate predicts transition from paroxysmal to long-
Int J Cardiol 255: 61–68, 2018. doi:10.1016/j.ijcard.2017.12.050. term persistent atrial fibrillation. Circulation 129: 1472–1482, 2014.
doi:10.1161/CIRCULATIONAHA.113.004742.
872. Li N, Timofeyev V, Tuteja D, Xu D, Lu L, Zhang Q, Zhang Z,
Singapuri A, Albert TR, Rajagopal AV, Bond CT, Periasamy M, 885. Wu MS, Gabriels J, Khan M, Shaban N, D’Amato SA, Liu CF,
Adelman J, Chiamvimonvat N. Ablation of a Ca21-activated K1 Markowitz SM, Ip JE, Thomas G, Singh P, Lerman BB, Patel A,
channel (SK2 channel) results in action potential prolongation in Cheung JW. Left atrial thrombus despite continuous direct oral anti-
atrial myocytes and atrial fibrillation. J Physiol 587: 1087–1100, coagulant or warfarin therapy in patients with atrial fibrillation:
2009. doi:10.1113/jphysiol.2008.167718. insights into rates and timing of thrombus resolution. J Interv Card
Electrophysiol 53: 159–167, 2018. doi:10.1007/s10840-018-0432-1.
873. Bonilla IM, Long VP 3rd, Vargas-Pinto P, Wright P, Belevych A, Lou
Q, Mowrey K, Yoo J, Binkley PF, Fedorov VV, Gyo € rke S, Janssen 886. Kannel WB, Abbott RD, Savage DD, McNamara PM. Epidemiologic
PM, Kilic A, Mohler PJ, Carnes CA. Calcium-activated potassium features of chronic atrial fibrillation: the Framingham study. N Engl
current modulates ventricular repolarization in chronic heart failure. J Med 306: 1018–1022, 1982. doi:10.1056/NEJM198204293061703.
PLoS One 9: e108824, 2014. doi:10.1371/journal.pone.0108824.
887. Dobrev D. Atrial Ca21/calmodulin-dependent protein kinase II: a
874. Yi F, Ling TY, Lu T, Wang XL, Li J, Claycomb WC, Shen WK, Lee HC. druggable master switch of atrial fibrillation-associated atrial
Down-regulation of the small conductance calcium-activated potas- remodeling? Heart Rhythm 16: 1089–1090, 2019. doi:10.1016/j.
sium channels in diabetic mouse atria. J Biol Chem 290: 7016– hrthm.2019.02.002.
7026, 2015. doi:10.1074/jbc.M114.607952. 888. Schotten U, Dobrev D, Platonov PG, Kottkamp H, Hindricks G.
875. Cerbai E, Sartiani L, DePaoli P, Pino R, Maccherini M, Bizzarri F, Current controversies in determining the main mechanisms of atrial
fibrillation. J Intern Med 279: 428–438, 2016. doi:10.1111/joim.
DiCiolla F, Davoli G, Sani G, Mugelli A. The properties of the pace-
12492.
maker current IF in human ventricular myocytes are modulated by
cardiac disease. J Mol Cell Cardiol 33: 441–448, 2001. doi:10.1006/ 889. Wijffels MC, Kirchhof CJ, Dorland R, Allessie MA. Atrial fibrillation
jmcc.2000.1316. begets atrial fibrillation. A study in awake chronically instrumented
goats. Circulation 92: 1954–1968, 1995. doi:10.1161/01.cir.92.7.1954.
876. Hoppe UC, Jansen E, S€ udkamp M, Beuckelmann DJ. Hyperpolariza-
tion-activated inward current in ventricular myocytes from normal 890. Lin YK, Chen YA, Lee TI, Chen YC, Chen SA, Chen YJ. Aging modu-
and failing human hearts. Circulation 97: 55–65, 1998. doi:10.1161/01. lates the substrate and triggers remodeling in atrial fibrillation. Circ
cir.97.1.55. J 82: 1237–1244, 2018. doi:10.1253/circj.CJ-17-0242.

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1167


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

891. Pandit SV, Workman AJ. Atrial electrophysiological remodeling and insights from computational modeling. J Mol Cell Cardiol 79: 32–
fibrillation in heart failure. Clin Med Insights Cardiol 10: 41–46, 41, 2015. doi:10.1016/j.yjmcc.2014.10.018.
2016. doi:10.4137/CMC.S39713.
906. Wong KR, Trezise AE, Crozatier B, Vandenberg JI. Loss of the nor-
892. Poulet C, Wettwer E, Grunnet M, Jespersen T, Fabritz L, Matschke mal epicardial to endocardial gradient of cftr mRNA expression in
K, Knaut M, Ravens U. Late sodium current in human atrial cardio- the hypertrophied rabbit left ventricle. Biochem Biophys Res
myocytes from patients in sinus rhythm and atrial fibrillation. PLoS Commun 278: 144–149, 2000. doi:10.1006/bbrc.2000.3754.
One 10: e0131432, 2015. doi:10.1371/journal.pone.0131432.
907. Hobai IA, O’Rourke B. Enhanced Ca21-activated Na1-Ca21
893. Dobrev D, Wehrens XH. Calcium-mediated cellular triggered activ- exchange activity in canine pacing-induced heart failure. Circ Res
ity in atrial fibrillation. J Physiol 595: 4001–4008, 2017. doi:10.1113/ 87: 690–698, 2000. doi:10.1161/01.res.87.8.690.
JP273048.
908. Gorski PA, Ceholski DK, Hajjar RJ. Altered myocardial calcium cy-
894. Li D, Melnyk P, Feng J, Wang Z, Petrecca K, Shrier A, Nattel S. cling and energetics in heart failure–a rational approach for dis-
Effects of experimental heart failure on atrial cellular and ionic elec- ease treatment. Cell Metab 21: 183–194, 2015. doi:10.1016/j.cmet.
trophysiology. Circulation 101: 2631–2638, 2000. doi:10.1161/01. 2015.01.005.
CIR.101.22.2631.
909. Chang SL, Chen YC, Yeh YH, Lai YJ, Yeh HI, Lin CI, Lin YK, Lin YJ,
895. Chen WT, Chen YC, Hsieh MH, Huang SY, Kao YH, Chen YA, Lin Wu TJ, Huang YK, Chen SA, Chen YJ. Heart failure enhances
YK, Chen SA, Chen YJ. The uremic toxin indoxyl sulfate increases arrhythmogenesis in pulmonary veins. Clin Exp Pharmacol Physiol
pulmonary vein and atrial arrhythmogenesis. J Cardiovasc 38: 666–674, 2011. doi:10.1111/j.1440-1681.2011.05553.x.
Electrophysiol 26: 203–210, 2015. doi:10.1111/jce.12554. 910. Priebe L, Beuckelmann DJ. Simulation study of cellular electric
properties in heart failure. Circ Res 82: 1206–1223, 1998. doi:10.
896. Gutierrez A, Van Wagoner DR. Oxidant and inflammatory mecha-
1161/01.res.82.11.1206.
nisms and targeted therapy in atrial fibrillation: an update. J
Cardiovasc Pharmacol 66: 523–529, 2015. doi:10.1097/FJC. 911. Luo X, Lin H, Pan Z, Xiao J, Zhang Y, Lu Y, Yang B, Wang Z.
0000000000000313. Down-regulation of miR-1/miR-133 contributes to re-expression
of pacemaker channel genes HCN2 and HCN4 in hypertrophic
897. Mihm MJ, Yu F, Carnes CA, Reiser PJ, McCarthy PM, Van Wagoner
heart. J Biol Chem 283: 20045–20052, 2008. doi:10.1074/jbc.
DR, Bauer JA. Impaired myofibrillar energetics and oxidative injury
M801035200.
during human atrial fibrillation. Circulation 104: 174–180, 2001.
doi:10.1161/01.CIR.104.2.174. 912. Zicha S, Fernández-Velasco M, Lonardo G, L’Heureux N, Nattel S.
Sinus node dysfunction and hyperpolarization-activated (HCN)
898. Anderson ME. Oxidant stress promotes disease by activating channel subunit remodeling in a canine heart failure model.
CaMKII. J Mol Cell Cardiol 89: 160–167, 2015. doi:10.1016/j.yjmcc. Cardiovasc Res 66: 472–481, 2005. doi:10.1016/j.cardiores.2005.
2015.10.014. 02.011.
899. Liu Z, Finet JE, Wolfram JA, Anderson ME, Ai X, Donahue JK. 913. Verkerk AO, Wilders R, Coronel R, Ravesloot JH, Verheijck EE.
Calcium/calmodulin-dependent protein kinase II causes atrial struc- Ionic remodeling of sinoatrial node cells by heart failure.
tural remodeling associated with atrial fibrillation and heart failure. Circulation 108: 760–766, 2003. doi:10.1161/01.CIR.0000083719.
Heart Rhythm 16: 1080–1088, 2019. doi:10.1016/j.hrthm.2019. 51661.B9.
01.013.
914. Verkerk AO, Veldkamp MW, Baartscheer A, Schumacher CA,
900. Kjekshus J. Arrhythmias and mortality in congestive heart fail- € pping C, van Ginneken AC, Ravesloot JH. Ionic mechanism of
Klo
ure. Am J Cardiol 65: 42I–48I, 1990. doi:10.1016/0002-9149(90) delayed afterdepolarizations in ventricular cells isolated from
90125-k. human end-stage failing hearts. Circulation 104: 2728–2733, 2001.
doi:10.1161/hc4701.099577.
901. Akar FG, Spragg DD, Tunin RS, Kass DA, Tomaselli GF.
Mechanisms underlying conduction slowing and arrhythmogenesis 915. Yeh YH, Wakili R, Qi XY, Chartier D, Boknik P, Ka€a
€b S, Ravens U,
in nonischemic dilated cardiomyopathy. Circ Res 95: 717–725, Coutu P, Dobrev D, Nattel S. Calcium-handling abnormalities
2004. doi:10.1161/01.RES.0000144125.61927.1c. underlying atrial arrhythmogenesis and contractile dysfunction in
dogs with congestive heart failure. Circ Arrhythm Electrophysiol 1:
902. Chen Y, Wakili R, Xiao J, Wu CT, Luo X, Clauss S, Dawson K, Qi X, 93–102, 2008. doi:10.1161/CIRCEP.107.754788.
Naud P, Shi YF, Tardif JC, Ka €a€b S, Dobrev D, Nattel S. Detailed
characterization of microRNA changes in a canine heart failure 916. Teare D. Asymmetrical hypertrophy of the heart in young adults. Br
model: relationship to arrhythmogenic structural remodeling. J Mol Heart J 20: 1–8, 1958. doi:10.1136/hrt.20.1.1.
Cell Cardiol 77: 113–124, 2014. doi:10.1016/j.yjmcc.2014.10.001.
917. Maron BJ, Gardin JM, Flack JM, Gidding SS, Kurosaki TT, Bild DE.
903. Li GR, Lau CP, Ducharme A, Tardif JC, Nattel S. Transmural action Prevalence of hypertrophic cardiomyopathy in a general popula-
potential and ionic current remodeling in ventricles of failing canine tion of young adults. Echocardiographic analysis of 4111 subjects in
hearts. Am J Physiol Heart Circ Physiol 283: H1031–H1041, 2002. the CARDIA Study. Coronary Artery Risk Development in (Young)
doi:10.1152/ajpheart.00105.2002. Adults. Circulation 92: 785–789, 1995. doi:10.1161/01.cir.92.4.785.

904. Kodama M, Kato K, Hirono S, Okura Y, Hanawa H, Yoshida T, 918. Gersh BJ, Maron BJ, Bonow RO, Dearani JA, Fifer MA, Link MS,
Hayashi M, Tachikawa H, Kashimura T, Watanabe K, Aizawa Y. Naidu SS, Nishimura RA, Ommen SR, Rakowski H, Seidman CE,
Linkage between mechanical and electrical alternans in patients Towbin JA, Udelson JE, Yancy CW; American College of
with chronic heart failure. J Cardiovasc Electrophysiol 15: 295– Cardiology Foundation/American Heart Association Task Force on
299, 2004. doi:10.1046/j.1540-8167.2004.03016.x. Practice Guidelines. American College of Cardiology Foundation/
American Heart Association Task Force on Practice Guidelines.
905. Nivala M, Song Z, Weiss JN, Qu Z. T-tubule disruption promotes 2011 ACCF/AHA Guideline for the Diagnosis and Treatment of
calcium alternans in failing ventricular myocytes: mechanistic Hypertrophic Cardiomyopathy: a report of the American College of

1168 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

Cardiology Foundation/American Heart Association Task Force on 931. Ferrantini C, Pioner JM, Mazzoni L, Gentile F, Tosi B, Rossi A,
Practice Guidelines. J Am Coll Cardiol 58: e212–e260, 2011. Belardinelli L, Tesi C, Palandri C, Matucci R, Cerbai E, Olivotto I,
doi:10.1016/j.jacc.2011.06.011. Poggesi C, Mugelli A, Coppini R. Late sodium current inhibitors to
treat exercise-induced obstruction in hypertrophic cardiomyopathy:
919. Maron BJ. Hypertrophic cardiomyopathy and other causes of sud- an in vitro study in human myocardium. Br J Pharmacol 175: 2635–
den cardiac death in young competitive athletes, with considera- 2652, 2018. doi:10.1111/bph.14223.
tions for preparticipation screening and criteria for disqualification.
Cardiol Clin 25: 399–414, 2007. doi:10.1016/j.ccl.2007.07.006. 932. Maron BJ, Pelliccia A. The heart of trained athletes: cardiac remod-
eling and the risks of sports, including sudden death. Circulation
920. Wigle ED, Rakowski H, Kimball BP, Williams WG. Hypertrophic car-
114: 1633–1644, 2006. doi:10.1161/CIRCULATIONAHA.106.613562.
diomyopathy. Clinical spectrum and treatment. Circulation 92:
1680–1692, 1995. doi:10.1161/01.cir.92.7.1680. 933. D’Souza A, Bucchi A, Johnsen AB, Logantha SJ, Monfredi O, Yanni
J, Prehar S, Hart G, Cartwright E, Wisloff U, Dobryznski H,
921. Decker JA, Rossano JW, Smith EO, Cannon B, Clunie SK, Gates C,
DiFrancesco D, Morris GM, Boyett MR. Exercise training reduces
Jefferies JL, Kim JJ, Price JF, Dreyer WJ, Towbin JA, Denfield SW.
resting heart rate via downregulation of the funny channel HCN4.
Risk factors and mode of death in isolated hypertrophic cardiomy-
Nat Commun 5: 3775, 2014. doi:10.1038/ncomms4775.
opathy in children. J Am Coll Cardiol 54: 250–254, 2009. doi:
10.1016/j.jacc.2009.03.051. 934. Atchley AE Jr, Douglas PS. Left ventricular hypertrophy in athletes:
922. Kessler EL, Boulaksil M, van Rijen HV, Vos MA, van Veen TA. morphologic features and clinical correlates. Cardiol Clin 25: 371–
Passive ventricular remodeling in cardiac disease: focus on hetero- 382, 2007. doi:10.1016/j.ccl.2007.06.009.
geneity. Front Physiol 5: 482, 2014. doi:10.3389/fphys.2014.00482. 935. Corrado D, Michieli P, Basso C, Schiavon M, Thiene G. How to
923. Maron BJ, Anan TJ, Roberts WC. Quantitative analysis of the distri- screen athletes for cardiovascular diseases. Cardiol Clin 25: 391–
bution of cardiac muscle cell disorganization in the left ventricular 397, 2007. doi:10.1016/j.ccl.2007.07.008.
wall of patients with hypertrophic cardiomyopathy. Circulation 63:
936. Lengyel C, Orosz A, Hegyi P, Komka Z, Udvardy A, Bosnyák E,
882–894, 1981. doi:10.1161/01.cir.63.4.882.
 th M, Wittmann T, Papp JG, Varro
Trájer E, Pavlik G, To  A, Baczko
 I.
924. Sotgia B, Sciagrá R, Olivotto I, Casolo G, Rega L, Betti I, Pupi A, Increased short-term variability of the QT interval in professional
Camici PG, Cecchi F. Spatial relationship between coronary micro- soccer players: possible implications for arrhythmia prediction.
vascular dysfunction and delayed contrast enhancement in PLoS One 6: e18751, 2011. doi:10.1371/journal.pone.0018751.
patients with hypertrophic cardiomyopathy. J Nucl Med 49: 1090–
937. Rowland T. Is the ‘athlete’s heart’ arrhythmogenic? Implications for
1096, 2008. doi:10.2967/jnumed.107.050138.
sudden cardiac death. Sports Med 41: 401–411, 2011. doi:10.2165/
925. Galati G, Leone O, Pasquale F, Olivotto I, Biagini E, Grigioni F, Pilato 11583940-000000000-00000.
E, Lorenzini M, Corti B, Foà A, Agostini V, Cecchi F, Rapezzi C.
Histological and histometric characterization of myocardial fibrosis 938. Tse G, Lai ET, Tse V, Yeo JM. Molecular and electrophysiological
in end-stage hypertrophic cardiomyopathy: a clinical-pathological mechanisms underlying cardiac arrhythmogenesis in diabetes mel-
study of 30 explanted hearts. Circ Heart Fail 9: e003090, 2016. litus. J Diabetes Res 2016: 2848759, 2016. doi:10.1155/2016/
doi:10.1161/CIRCHEARTFAILURE.116.003090. 2848759.

926. Hurtado-de-Mendoza D, Corona-Villalobos CP, Pozios I, Gonzales 939. Zhang Y, Xiao J, Lin H, Luo X, Wang H, Bai Y, Wang J, Zhang H,
J, Soleimanifard Y, Sivalokanathan S, Montoya-Cerrillo D, Vakrou S, Yang B, Wang Z. Ionic mechanisms underlying abnormal QT pro-
Kamel I, Mormontoy-Laurel W, Dolores-Cerna K, Suarez J, Perez- longation and the associated arrhythmias in diabetic rabbits: a role
Melo S, Bluemke DA, Abraham TP, Zimmerman SL, Abraham MR. of rapid delayed rectifier K1 current. Cell Physiol Biochem 19:
Diffuse interstitial fibrosis assessed by cardiac magnetic resonance 225–238, 2007. doi:10.1159/000100642.
is associated with dispersion of ventricular repolarization in
940. Fu X, Pan Y, Cao Q, Li B, Wang S, Du H, Duan N, Li X. Metformin
patients with hypertrophic cardiomyopathy. J Arrhythm 33: 201–
restores electrophysiology of small conductance calcium-activated
207, 2017. doi:10.1016/j.joa.2016.10.005.
potassium channels in the atrium of GK diabetic rats. BMC
927. Cannon RO 3rd, Schenke WH, Bonow RO, Leon MB, Rosing DR. Cardiovasc Disord 18: 63, 2018. doi:10.1186/s12872-018-0805-5.
Left ventricular pulsus alternans in patients with hypertrophic car-
diomyopathy and severe obstruction to left ventricular outflow. 941. D’Souza A, Howarth FC, Yanni J, Dobryznski H, Boyett MR,
Circulation 73: 276–285, 1986. doi:10.1161/01.cir.73.2.276. Adeghate E, Bidasee KR, Singh J. Left ventricle structural remodel-
ling in the prediabetic Goto-Kakizaki rat. Exp Physiol 96: 875–888,
928. Maurizi N, Passantino S, Spaziani G, Girolami F, Arretini A, Targetti 2011. doi:10.1113/expphysiol.2011.058271.
M, Pollini I, Tomberli A, Pradella S, Calabri GB, Vinattieri V,
Bertaccini B, Leone O, De Simone L, Rapezzi C, Marchionni N, 942. Lin YC, Huang J, Kan H, Castranova V, Frisbee JC, Yu HG.
Cecchi F, Favilli S, Olivotto I. Long-term outcomes of pediatric- Defective calcium inactivation causes long QT in obese insulin-re-
onset hypertrophic cardiomyopathy and age-specific risk factors sistant rat. Am J Physiol Heart Circ Physiol 302: H1013–H1022,
for lethal arrhythmic events. JAMA Cardiol 3: 520–525, 2018. 2012. doi:10.1152/ajpheart.00837.2011.
doi:10.1001/jamacardio.2018.0789.
943. Nygren A, Olson ML, Chen KY, Emmett T, Kargacin G, Shimoni Y.
929. Adabag AS, Casey SA, Kuskowski MA, Zenovich AG, Maron BJ. Propagation of the cardiac impulse in the diabetic rat heart:
Spectrum and prognostic significance of arrhythmias on ambula- reduced conduction reserve. J Physiol 580: 543–560, 2007.
tory Holter electrocardiogram in hypertrophic cardiomyopathy. J doi:10.1113/jphysiol.2006.123729.
Am Coll Cardiol 45: 697–704, 2005. doi:10.1016/j.jacc.2004.11.043.
944. Fowlkes V, Clark J, Fix C, Law BA, Morales MO, Qiao X, Ako-Asare
930. Ulus T, Kudaiberdieva G, Gorenek B. The onset mechanisms of K, Goldsmith JG, Carver W, Murray DB, Goldsmith EC. Type II diabe-
ventricular tachycardia. Int J Cardiol 167: 619–623, 2013. tes promotes a myofibroblast phenotype in cardiac fibroblasts. Life
doi:10.1016/j.ijcard.2012.09.034. Sci 92: 669–676, 2013. doi:10.1016/j.lfs.2013.01.003.

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1169


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

945. Ghaly HA, Boyle PM, Vigmond EJ, Shimoni Y, Nygren A. porcine heart. Circulation 56: 217–224, 1977. doi:10.1161/01.
Simulations of reduced conduction reserve in the diabetic rat heart: cir.56.2.217.
response to uncoupling and reduced excitability. Ann Biomed Eng
38: 1415–1425, 2010. doi:10.1007/s10439-009-9855-2. ber AG. Resting membrane potential, extracellular potassium
961. Kle
activity, and intracellular sodium activity during acute global ische-
946. Shao CH, Rozanski GJ, Patel KP, Bidasee KR. Dyssynchronous mia in isolated perfused guinea pig hearts. Circ Res 52: 442–450,
(non-uniform) Ca21 release in myocytes from streptozotocin- 1983. doi:10.1161/01.RES.52.4.442.
induced diabetic rats. J Mol Cell Cardiol 42: 234–246, 2007.
doi:10.1016/j.yjmcc.2006.08.018. 962. Ferrero JM Jr, Sa´iz J, Ferrero JM, Thakor NV. Simulation of action
potentials from metabolically impaired cardiac myocytes. Role of
947. Shao CH, Tian C, Ouyang S, Moore CJ, Alomar F, Nemet I, D'Souza ATP-sensitive K1 current. Circ Res 79: 208–221, 1996. doi:10.1161/
A, Nagai R, Kutty S, Rozanski GJ, Ramanadham S, Singh J, Bidasee 01.RES.79.2.208.
KR. Carbonylation induces heterogeneity in cardiac ryanodine re-
ceptor function in diabetes mellitus. Mol Pharmacol 82: 383–399, 963. Beardslee MA, Lerner DL, Tadros PN, Laing JG, Beyer EC, Yamada
2012. doi:10.1124/mol.112.078352. KA, Kleber AG, Schuessler RB, Saffitz JE. Dephosphorylation and
intracellular redistribution of ventricular connexin43 during electri-
948. Yaras N, Ugur M, Ozdemir S, Gurdal H, Purali N, Lacampagne A,
cal uncoupling induced by ischemia. Circ Res 87: 656–662, 2000.
Vassort G, Turan B. Effects of diabetes on ryanodine receptor Ca
doi:10.1161/01.res.87.8.656.
release channel (RyR2) and Ca21 homeostasis in rat heart.
Diabetes 54: 3082–3088, 2005. doi:10.2337/diabetes.54.11.3082. 964. Janse MJ, Kleber AG. Electrophysiological changes and ventricular
arrhythmias in the early phase of regional myocardial ischemia.
949. Netticadan T, Temsah RM, Kent A, Elimban V, Dhalla NS.
Circ Res 49: 1069–1081, 1981. doi:10.1161/01.res.49.5.1069.
Depressed levels of Ca21-cycling proteins may underlie sarcoplas-
mic reticulum dysfunction in the diabetic heart. Diabetes 50: 2133– 965. McCallister LP, Trapukdi S, Neely JR. Morphometric observations
2138, 2001. doi:10.2337/diabetes.50.9.2133. on the effects of ischemia in the isolated perfused rat heart. J Mol
950. Tuncay E, Okatan EN, Toy A, Turan B. Enhancement of cellular anti- Cell Cardiol 11: 619–630, 1979. doi:10.1016/0022-2828(79)90376-6.
oxidant-defence preserves diastolic dysfunction via regulation of ber AG, Janse MJ, Wilms-Schopmann FJ, Wilde AA, Coronel R.
966. Kle
both diastolic Zn21 and Ca21 and prevention of RyR2-leak in Changes in conduction velocity during acute ischemia in ventricular
hyperglycemic cardiomyocytes. Oxid Med Cell Longev 2014:
myocardium of the isolated porcine heart. Circulation 73: 189–198,
290381, 2014. doi:10.1155/2014/290381.
1986. doi:10.1161/01.cir.73.1.189.
951. Hamilton S, Terentyev D. Proarrhythmic remodeling of calcium ho-
 I, Husti Z, Lang V, Leprán I, Light PE. Sarcolemmal KATP
967. Baczko
meostasis in cardiac disease; implications for diabetes and obesity.
channel modulators and cardiac arrhythmias. Curr Med Chem 18:
Front Physiol 9: 1517, 2018. doi:10.3389/fphys.2018.01517.
3640–3661, 2011. doi:10.2174/092986711796642472.
952. Okatan EN, Durak AT, Turan B. Electrophysiological basis of meta-
968. Kaplinsky E, Ogawa S, Balke CW, Dreifus LS. Two periods of early
bolic-syndrome-induced cardiac dysfunction. Can J Physiol
Pharmacol 94: 1064–1073, 2016. doi:10.1139/cjpp-2015-0531. ventricular arrhythmia in the canine acute myocardial infarction
model. Circulation 60: 397–403, 1979. doi:10.1161/01.cir.60.2.397.
953. Nikolic G, Bishop RL, Singh JB. Sudden death recorded during
Holter monitoring. Circulation 66: 218–225, 1982. doi:10.1161/01. 969. Martinez-Navarro H, Minchole  A, Bueno-Orovio A, Rodriguez B.
cir.66.1.218. High arrhythmic risk in antero-septal acute myocardial ischemia is
explained by increased transmural reentry occurrence. Sci Rep 9:
954. Pantridge JF, Webb SW, Adgey AA. Arrhythmias in the first hours of 16803, 2019. doi:10.1038/s41598-019-53221-2.
acute myocardial infarction. Prog Cardiovasc Dis 23: 265–278,
1981. doi:10.1016/0033-0620(81)90016-5. 970. Scho€ mig A, Dart AM, Dietz R, Mayer E, K€
ubler W. Release of endog-
enous catecholamines in the ischemic myocardium of the rat. Part
955. Manning AS, Hearse DJ. Reperfusion-induced arrhythmias: mecha- A: Locally mediated release. Circ Res 55: 689–701, 1984. doi:10.
nisms and prevention. J Mol Cell Cardiol 16: 497–518, 1984. 1161/01.res.55.5.689.
doi:10.1016/s0022-2828(84)80638-0.
971. Arnsdorf MF, Sawicki GJ. The effects of lysophosphatidylcholine, a
956. Tzivoni D, Keren A, Granot H, Gottlieb S, Benhorin J, Stern S. toxic metabolite of ischemia, on the components of cardiac excit-
Ventricular fibrillation caused by myocardial reperfusion in ability in sheep Purkinje fibers. Circ Res 49: 16–30, 1981. doi:10.1161/
Prinzmetal’s angina. Am Heart J 105: 323–325, 1983. doi:10.1016/ 01.res.49.1.16.
0002-8703(83)90534-3.
972. Coronel R, Wilms-Schopman FJ, Opthof T, van Capelle FJ, Janse
957. Maseri A, Severi S, Marzullo P. Role of coronary arterial spasm in MJ. Injury current and gradients of diastolic stimulation threshold,
sudden coronary ischemic death. Ann NY Acad Sci 382: 204–217,
TQ potential, and extracellular potassium concentration during
1982. doi:10.1111/j.1749-6632.1982.tb55218.x.
acute regional ischemia in the isolated perfused pig heart. Circ Res
958. Clark BB, Cummings JR. Arrhythmias following experimental coro- 68: 1241–1249, 1991. doi:10.1161/01.res.68.5.1241.
nary occlusion and their response to drugs. Ann NY Acad Sci 64:
973. Janse MJ, van Capelle FJ. Electrotonic interactions across an inex-
543–551, 1956. doi:10.1111/j.1749-6632.1956.tb36828.x.
citable region as a cause of ectopic activity in acute regional myo-
959. Kabell G, Scherlag BJ, Hope RR, Lazzara R. Regional myocardial cardial ischemia. A study in intact porcine and canine hearts and
blood flow and ventricular arrhythmias following one-stage and computer models. Circ Res 50: 527–537, 1982. doi:10.1161/01.
two-stage coronary artery occlusion in anesthetized dogs. Am res.50.4.527.
Heart J 104: 537–544, 1982. doi:10.1016/0002-8703(82)90224-1.
974. Katzung BG, Hondeghem LM, Grant AO. Letter: Cardiac ventricular
960. Downar E, Janse MJ, Durrer D. The effect of acute coronary artery automaticity induced by current of injury. Pflugers Arch 360: 193–
occlusion on subepicardial transmembrane potentials in the intact 197, 1975. doi:10.1007/BF00580542.

1170 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

975. Qin D, Zhang ZH, Caref EB, Boutjdir M, Jain P, el-Sherif N. Cellular 989. Ho KK, Pinsky JL, Kannel WB, Levy D. The epidemiology of heart
and ionic basis of arrhythmias in postinfarction remodeled ventricu- failure: the Framingham Study. J Am Coll Cardiol 22: 6A–13A,
lar myocardium. Circ Res 79: 461–473, 1996. doi:10.1161/01.res.79. 1993. doi:10.1016/0735-1097(93)90455-a.
3.461.
990. Benjamin EJ, Virani SS, Callaway CW, Chamberlain AM, Chang AR,
976. Friedman PL, Fenoglio JJ, Wit AL. Time course for reversal of elec- Cheng S, et al. Heart Disease and Stroke Statistics-2018 Update: A
trophysiological and ultrastructural abnormalities in subendocardial Report From the American Heart Association. Circulation 137:
Purkinje fibers surviving extensive myocardial infarction in dogs. e67–e492, 2018. doi:10.1161/CIR.0000000000000558.
Circ Res 36: 127–144, 1975. doi:10.1161/01.res.36.1.127.
991. Fishman GI, Chugh SS, Dimarco JP, Albert CM, Anderson ME,
977. Lue WM, Boyden PA. Abnormal electrical properties of myocytes Bonow RO, Buxton AE, Chen PS, Estes M, Jouven X, Kwong R,
from chronically infarcted canine heart. Alterations in Vmax and the Lathrop DA, Mascette AM, Nerbonne JM, O’Rourke B, Page RL,
transient outward current. Circulation 85: 1175–1188, 1992. doi:10. Roden DM, Rosenbaum DS, Sotoodehnia N, Trayanova NA,
1161/01.cir.85.3.1175. Zheng ZJ. Sudden cardiac death prediction and prevention:
report from a National Heart, Lung, and Blood Institute and Heart
978. Pinto JM, Boyden PA. Reduced inward rectifying and increased Rhythm Society Workshop. Circulation 122: 2335–2348, 2010.
E-4031-sensitive K1 current density in arrhythmogenic suben- doi:10.1161/CIRCULATIONAHA.110.976092.
docardial purkinje myocytes from the infarcted heart. J
Cardiovasc Electrophysiol 9: 299–311, 1998. doi:10.1111/j.1540- 992. Mirza M, Strunets A, Shen WK, Jahangir A. Mechanisms of arrhyth-
8167.1998.tb00915.x. mias and conduction disorders in older adults. Clin Geriatr Med
28: 555–573, 2012. doi:10.1016/j.cger.2012.08.005.
979. Boyden PA, Pinto JM. Reduced calcium currents in subendocardial
Purkinje myocytes that survive in the 24- and 48-hour infarcted 993. Davies CH, Ferrara N, Harding SE. Beta-adrenoceptor function
heart. Circulation 89: 2747–2759, 1994. doi:10.1161/01.cir.89.6. changes with age of subject in myocytes from non-failing human
2747. ventricle. Cardiovasc Res 31: 152–156, 1996. doi:10.1016/S0008-
6363(95)00187-5.
980. Jeck C, Pinto J, Boyden P. Transient outward currents in subendo-
994. Gazoti Debessa CR, Mesiano Maifrino LB, Rodrigues de Souza R.
cardial Purkinje myocytes surviving in the infarcted heart.
Age related changes of the collagen network of the human heart.
Circulation 92: 465–473, 1995. doi:10.1161/01.CIR.92.3.465.
Mech Ageing Dev 122: 1049–1058, 2001. doi:10.1016/s0047-6374
981. Cabo C, Boyden PA. Electrical remodeling of the epicardial border (01)00238-x.
zone in the canine infarcted heart: a computational analysis. Am J
995. Lakatta EG, Levy D. Arterial and cardiac aging: major shareholders
Physiol Heart Circ Physiol 284: H372–H384, 2003. doi:10.1152/
in cardiovascular disease enterprises: Part II: the aging heart in
ajpheart.00512.2002.
health: links to heart disease. Circulation 107: 346–354, 2003.
982. Pu J, Boyden PA. Alterations of Na1 currents in myocytes from epi- doi:10.1161/01.cir.0000048893.62841.f7.
cardial border zone of the infarcted heart. A possible ionic mecha-
996. Lakatta EG. Cardiovascular regulatory mechanisms in advanced
nism for reduced excitability and postrepolarization refractoriness.
age. Physiol Rev 73: 413–467, 1993. doi:10.1152/physrev.1993.73.2.
Circ Res 81: 110–119, 1997. doi:10.1161/01.RES.81.1.110.
413.
983. Peters NS, Coromilas J, Severs NJ, Wit AL. Disturbed connexin43
997. Larson ED, St Clair JR, Sumner WA, Bannister RA, Proenza C.
gap junction distribution correlates with the location of reentrant
Depressed pacemaker activity of sinoatrial node myocytes contrib-
circuits in the epicardial border zone of healing canine infarcts that utes to the age-dependent decline in maximum heart rate. Proc
cause ventricular tachycardia. Circulation 95: 988–996, 1997. Natl Acad Sci USA 110: 18011–18016, 2013. doi:10.1073/pnas.
doi:10.1161/01.CIR.95.4.988. 1308477110.
984. Dillon SM, Allessie MA, Ursell PC, Wit AL. Influences of anisotropic 998. Tellez JO, McZewski M, Yanni J, Sutyagin P, Mackiewicz U,
tissue structure on reentrant circuits in the epicardial border zone Atkinson A, Inada S, Beresewicz A, Billeter R, Dobrzynski H, Boyett
of subacute canine infarcts. Circ Res 63: 182–206, 1988. MR. Ageing-dependent remodelling of ion channel and Ca21 clock
doi:10.1161/01.res.63.1.182. genes underlying sino-atrial node pacemaking. Exp Physiol 96:
1163–1178, 2011. doi:10.1113/expphysiol.2011.057752.
985. Gardner PI, Ursell PC, Fenoglio JJ Jr, Wit AL. Electrophysiologic
and anatomic basis for fractionated electrograms recorded from 999. Schmidlin O, Bharati S, Lev M, Schwartz JB. Effects of physiological
healed myocardial infarcts. Circulation 72: 596–611, 1985. aging on cardiac electrophysiology in perfused Fischer 344 rat
doi:10.1161/01.cir.72.3.596. hearts. Am J Physiol Heart Circ Physiol 262: H97–H105, 1992.
doi:10.1152/ajpheart.1992.262.1.H97.
986. Ursell PC, Gardner PI, Albala A, Fenoglio JJ Jr, Wit AL. Structural
and electrophysiological changes in the epicardial border zone of 1000. Monfredi O, Boyett MR. Sick sinus syndrome and atrial fibrillation in
canine myocardial infarcts during infarct healing. Circ Res 56: 436– older persons—a view from the sinoatrial nodal myocyte. J Mol
451, 1985. doi:10.1161/01.res.56.3.436. Cell Cardiol 83: 88–100, 2015. doi:10.1016/j.yjmcc.2015.02.003.

987. Aggarwal R, Boyden PA. Diminished Ca21 and Ba21 currents in 1001. Bonda TA, Szynaka B, Sokołowska M, Dziemidowicz M, Winnicka
myocytes surviving in the epicardial border zone of the 5-day MM, Chyczewski L, Kamin ski KA. Remodeling of the intercalated
infarcted canine heart. Circ Res 77: 1180–1191, 1995. doi:10.1161/01. disc related to aging in the mouse heart. J Cardiol 68: 261–268,
res.77.6.1180. 2016. doi:10.1016/j.jjcc.2015.10.001.

988. Tomek J, Bub G. Hypertension-induced remodelling: on the inter- 1002. Jones SA. Ageing to arrhythmias: conundrums of connections in
actions of cardiac risk factors. J Physiol 595: 4027–4036, 2017. the ageing heart. J Pharm Pharmacol 58: 1571–1576, 2006.
doi:10.1113/JP273043. doi:10.1211/jpp.58.12.0002.

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1171


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

1003. Lie JT, Hammond PI. Pathology of the senescent heart: anatomic 1017. Ferrara N, Komici K, Corbi G, Pagano G, Furgi G, Rengo C,
observations on 237 autopsy studies of patients 90 to 105 years Femminella GD, Leosco D, Bonaduce D. Beta-adrenergic receptor
old. Mayo Clin Proc 63: 552–564, 1988. doi:10.1016/s0025-6196 responsiveness in aging heart and clinical implications. Front
(12)64885-x. Physiol 4: 396, 2014. doi:10.3389/fphys.2013.00396.

1004. Anversa P, Palackal T, Sonnenblick EH, Olivetti G, Meggs LG, 1018. Xiao RP, Tomhave ED, Wang DJ, Ji X, Boluyt MO, Cheng H, Lakatta
Capasso JM. Myocyte cell loss and myocyte cellular hyperplasia in EG, Koch WJ. Age-associated reductions in cardiac beta1- and
the hypertrophied aging rat heart. Circ Res 67: 871–885, 1990. beta2-adrenergic responses without changes in inhibitory G pro-
doi:10.1161/01.res.67.4.871. teins or receptor kinases. J Clin Invest 101: 1273–1282, 1998.
doi:10.1172/JCI1335.
1005. Gottwald M, Gottwald E, Dhein S. Age-related electrophysiological
and histological changes in rabbit hearts: age-related changes in 1019. Terrenoire C, Clancy CE, Cormier JW, Sampson KJ, Kass RS.
electrophysiology. Int J Cardiol 62: 97–106, 1997. doi:10.1016/ Autonomic control of cardiac action potentials: role of potassium
s0167-5273(97)00183-6. channel kinetics in response to sympathetic stimulation. Circ Res
96: e25–e34, 2005. doi:10.1161/01.RES.0000160555.58046.9a.
1006. Olivetti G, Melissari M, Capasso JM, Anversa P. Cardiomyopathy of
the aging human heart. Myocyte loss and reactive cellular hypertro- 1020. Hartzell HC, Mery PF, Fischmeister R, Szabo G. Sympathetic regula-
phy. Circ Res 68: 1560–1568, 1991. doi:10.1161/01.res.68.6.1560. tion of cardiac calcium current is due exclusively to cAMP-depend-
ent phosphorylation. Nature 351: 573–576, 1991. doi:10.1038/
1007. Dibb KM, Rueckschloss U, Eisner DA, Isenberg G, Trafford AW. 351573a0.
Mechanisms underlying enhanced cardiac excitation contraction
coupling observed in the senescent sheep myocardium. J Mol Cell 1021. Korkushko OV, Shatilo VB, Plachinda Yu I, Shatilo TV. Autonomic
Cardiol 37: 1171–1181, 2004. doi:10.1016/j.yjmcc.2004.09.005. control of cardiac chronotropic function in man as a function of
age: assessment by power spectral analysis of heart rate variability.
1008. Sorrentino A, Signore S, Qanud K, Borghetti G, Meo M, Cannata A, J Auton Nerv Syst 32: 191–198, 1991. doi:10.1016/0165-1838(91)
Zhou Y, Wybieralska E, Luciani M, Kannappan R, Zhang E, Matsuda 90113-H.
A, Webster A, Cimini M, Kertowidjojo E, D’Alessandro DA,
Wunimenghe O, Michler RE, Royer C, Goichberg P, Leri A, Barrett €nicke K, Brodde OE. Age-depend-
1022. Poller U, Nedelka G, Radke J, Po
EG, Anversa P, Hintze TH, Rota M. Myocyte repolarization modu- ent changes in cardiac muscarinic receptor function in healthy vol-
lates myocardial function in aging dogs. Am J Physiol Heart Circ unteers. J Am Coll Cardiol 29: 187–193, 1997. doi:10.1016/S0735-
Physiol 310: H873–H890, 2016. doi:10.1152/ajpheart.00682.2015. 1097(96)00437-8.

1009. Josephson IR, Guia A, Stern MD, Lakatta EG. Alterations in proper- 1023. Kalla M, Herring N, Paterson DJ. Cardiac sympatho-vagal balance
ties of L-type Ca channels in aging rat heart. J Mol Cell Cardiol 34: and ventricular arrhythmia. Auton Neurosci 199: 29–37, 2016.
297–308, 2002. doi:10.1006/jmcc.2001.1512. doi:10.1016/j.autneu.2016.08.016.

1010. Signore S, Sorrentino A, Borghetti G, Cannata A, Meo M, Zhou Y, 1024. Sesti F, Abbott GW, Wei J, Murray KT, Saksena S, Schwartz PJ,
Kannappan R, Pasqualini F, O'Malley H, Sundman M, Tsigkas N, Priori SG, Roden DM, George AL Jr, Goldstein SA. A common poly-
Zhang E, Arranto C, Mangiaracina C, Isobe K, Sena BF, Kim J, morphism associated with antibiotic-induced cardiac arrhythmia.
Goichberg P, Nahrendorf M, Isom LL, Leri A, Anversa P, Rota M. Proc Natl Acad Sci USA 97: 10613–10618, 2000. doi:10.1073/
Late Na1 current and protracted electrical recovery are critical pnas.180223197.
determinants of the aging myopathy. Nat Commun 6: 8803, 2015.
doi:10.1038/ncomms9803. 1025. Baruscotti M, Bucchi A, Milanesi R, Paina M, Barbuti A, Gnecchi-
Ruscone T, Bianco E, Vitali-Serdoz L, Cappato R, DiFrancesco D. A
1011. Koban MU, Moorman AF, Holtz J, Yacoub MH, Boheler KR. gain-of-function mutation in the cardiac pacemaker HCN4 channel
Expressional analysis of the cardiac Na-Ca exchanger in rat devel- increasing cAMP sensitivity is associated with familial Inappropriate
opment and senescence. Cardiovasc Res 37: 405–423, 1998. Sinus Tachycardia. Eur Heart J 38: 280–288, 2017. doi:10.1093/
doi:10.1016/S0008-6363(97)00276-9. eurheartj/ehv582.

1012. Lompre AM, Lambert F, Lakatta EG, Schwartz K. Expression of sar- 1026. Jenewein T, Neumann T, Erkapic D, Kuniss M, Verhoff MA, Thiel G,
coplasmic reticulum Ca21-ATPase and calsequestrin genes in rat Kauferstein S. Influence of genetic modifiers on sudden cardiac
heart during ontogenic development and aging. Circ Res 69: death cases. Int J Legal Med 132: 379–385, 2018. doi:10.1007/
1380–1388, 1991. doi:10.1161/01.res.69.5.1380. s00414-017-1739-7.

1013. Taffet GE, Tate CA. CaATPase content is lower in cardiac sarco- 1027. Rudy Y. Mathematical modeling of complex biological systems:
plasmic reticulum isolated from old rats. Am J Physiol Heart Circ from genes and molecules to organs and organisms: heart. In:
Physiol 264: H1609–H1614, 1993. doi:10.1152/ajpheart.1993.264.5. Comprehensive Biophysics. Simulation and Modeling, edited by
H1609. Weinstein H. Oxford: Academic Press, 2012.

1014. Xu A, Narayanan N. Effects of aging on sarcoplasmic reticulum 1028. Vaidyanathan R, Reilly L, Eckhardt LL. Caveolin-3 microdomain: ar-
Ca21-cycling proteins and their phosphorylation in rat myocardium. rhythmia implications for potassium inward rectifier and cardiac so-
Am J Physiol Heart Circ Physiol 275: H2087–H2094, 1998. dium channel. Front Physiol 9: 1548, 2018. doi:10.3389/fphys.
doi:10.1152/ajpheart.1998.275.6.H2087. 2018.01548.

1015. Zhou YY, Lakatta EG, Xiao RP. Age-associated alterations in cal- 1029. Zhang Q, Chen J, Qin Y, Wang J, Zhou L. Mutations in voltage-
cium current and its modulation in cardiac myocytes. Drugs Aging gated L-type calcium channel: implications in cardiac arrhythmia.
13: 159–171, 1998. doi:10.2165/00002512-199813020-00007. Channels (Austin) 12: 201–218, 2018. doi:10.1080/19336950.
2018.1499368.
1016. Stratton JR, Levy WC, Caldwell JH, Jacobson A, May J, Matsuoka
D, Madden K. Effects of aging on cardiovascular responses to para- 1030. Olesen MS, Bentzen BH, Nielsen JB, Steffensen AB, David JP,
sympathetic withdrawal. J Am Coll Cardiol 41: 2077–2083, 2003. Jabbari J, Jensen HK, Haunsø S, Svendsen JH, Schmitt N.
doi:10.1016/S0735-1097(03)00418-2. Mutations in the potassium channel subunit KCNE1 are associated

1172 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

with early-onset familial atrial fibrillation. BMC Med Genet 13: 24, primary arrhythmia syndromes. Europace 15: 1389–1406, 2013.
2012. doi:10.1186/1471-2350-13-24. ] doi:10.1093/europace/eut272.

1031. Restier L, Cheng L, Sanguinetti MC. Mechanisms by which atrial fi- 1045. Giustetto C, Schimpf R, Mazzanti A, Scrocco C, Maury P, Anttonen
brillation-associated mutations in the S1 domain of KCNQ1 slow O, Probst V, Blanc JJ, Sbragia P, Dalmasso P, Borggrefe M, Gaita F.
deactivation of IKs channels. J Physiol 586: 4179–4191, 2008. Long-term follow-up of patients with short QT syndrome. J Am Coll
doi:10.1113/jphysiol.2008.157511. Cardiol 58: 587–595, 2011. doi:10.1016/j.jacc.2011.03.038.

1032. Steffensen AB, Refsgaard L, Andersen MN, Vallet C, Mujezinovic A, 1046. Bellocq C, van Ginneken AC, Bezzina CR, Alders M, Escande D,
Haunsø S, Svendsen JH, Olesen SP, Olesen MS, Schmitt N. IKs Mannens MM, Baro  I, Wilde AA. Mutation in the KCNQ1 gene lead-
gain- and loss-of-function in early-onset lone atrial fibrillation. J ing to the short QT-interval syndrome. Circulation 109: 2394–
Cardiovasc Electrophysiol 26: 715–723, 2015. doi:10.1111/jce.12666. 2397, 2004. doi:10.1161/01.CIR.0000130409.72142.FE.

1033. Delaney JT, Muhammad R, Blair MA, Kor K, Fish FA, Roden DM, 1047. Brugada R, Hong K, Dumaine R, Cordeiro J, Gaita F, Borggrefe M,
Darbar D. A KCNJ8 mutation associated with early repolarization Menendez TM, Brugada J, Pollevick GD, Wolpert C, Burashnikov E,
and atrial fibrillation. Europace 14: 1428–1432, 2012. doi:10.1093/ Matsuo K, Wu YS, Guerchicoff A, Bianchi F, Giustetto C, Schimpf R,
europace/eus150. Brugada P, Antzelevitch C. Sudden death associated with short-QT
syndrome linked to mutations in HERG. Circulation 109: 30–35,
1034. Ishikawa T, Ohno S, Murakami T, Yoshida K, Mishima H, Fukuoka T, 2004. doi:10.1161/01.CIR.0000109482.92774.3A.
Kimoto H, Sakamoto R, Ohkusa T, Aiba T, Nogami A, Sumitomo N,
Shimizu W, Yoshiura KI, Horigome H, Horie M, Makita N. Sick sinus 1048. Priori SG, Pandit SV, Rivolta I, Berenfeld O, Ronchetti E, Dhamoon
syndrome with HCN4 mutations shows early onset and frequent A, Napolitano C, Anumonwo J, di Barletta MR, Gudapakkam S, Bosi
association with atrial fibrillation and left ventricular noncompac- G, Stramba-Badiale M, Jalife J. A novel form of short QT syndrome
tion. Heart Rhythm 14: 717–724, 2017. doi:10.1016/j.hrthm.2017. (SQT3) is caused by a mutation in the KCNJ2 gene. Circ Res 96:
01.020. 800–807, 2005. doi:10.1161/01.RES.0000162101.76263.8c.

1035. Krahn AD, Healey JS, Chauhan V, Birnie DH, Simpson CS, 1049. Templin C, Ghadri JR, Rougier JS, Baumer A, Kaplan V, Albesa M,
Champagne J, Gardner M, Sanatani S, Exner DV, Klein GJ, Yee R, Sticht H, Rauch A, Puleo C, Hu D, Barajas-Martinez H, Antzelevitch
Skanes AC, Gula LJ, Gollob MH. Systematic assessment of patients C, L€
uscher TF, Abriel H, Duru F. Identification of a novel loss-of-
with unexplained cardiac arrest: Cardiac Arrest Survivors With function calcium channel gene mutation in short QT syndrome
Preserved Ejection Fraction Registry (CASPER). Circulation 120: (SQTS6). Eur Heart J 32: 1077–1088, 2011. doi:10.1093/eurheartj/
278–285, 2009. doi:10.1161/CIRCULATIONAHA.109.853143. ehr076.

1050. Antzelevitch C, Pollevick GD, Cordeiro JM, Casis O, Sanguinetti


1036. Curran ME, Splawski I, Timothy KW, Vincent GM, Green ED, Keating
MC, Aizawa Y, Guerchicoff A, Pfeiffer R, Oliva A, Wollnik B, Gelber
MT. A molecular basis for cardiac arrhythmia: HERG mutations
P, Bonaros EP Jr, Burashnikov E, Wu Y, Sargent JD, Schickel S,
cause long QT syndrome. Cell 80: 795–803, 1995. doi:10.1016/
Oberheiden R, Bhatia A, Hsu LF, Haïssaguerre M, Schimpf R,
0092-8674(95)90358-5.
Borggrefe M, Wolpert C. Loss-of-function mutations in the cardiac
1037. Wang Q, Curran ME, Splawski I, Burn TC, Millholland JM, VanRaay calcium channel underlie a new clinical entity characterized by ST-
TJ, Shen J, Timothy KW, Vincent GM, de Jager T, Schwartz PJ, segment elevation, short QT intervals, and sudden cardiac death.
Toubin JA, Moss AJ, Atkinson DL, Landes GM, Connors TD, Circulation 115: 442–449, 2007. doi:10.1161/CIRCULATIONAHA.
Keating MT. Positional cloning of a novel potassium channel gene: 106.668392.
KVLQT1 mutations cause cardiac arrhythmias. Nat Genet 12: 17–23,
1051. Roussel J, Labarthe F, Thireau J, Ferro F, Farah C, Roy J, Horiuchi
1996. doi:10.1038/ng0196-17.
M, Tardieu M, Lefort B, Benoist J, Lacampagne A, Richard S,
1038. Antzelevitch C, Shimizu W. Cellular mechanisms underlying the Fauconnier J, Babuty D, Le Guennec JY. Carnitine deficiency indu-
long QT syndrome. Curr Opin Cardiol 17: 43–51, 2002. doi:10.1097/ ces a short QT syndrome. Heart Rhythm 13: 165–174, 2016.
00001573-200201000-00007. doi:10.1016/j.hrthm.2015.07.027.

1039. Sicouri S, Glass A, Ferreiro M, Antzelevitch C. Transseptal disper-  A, Tran TA, Fontanaud P, Bois P, Babuty D, Labarthe
1052. Ferro F, Ouille
sion of repolarization and its role in the development of torsade de F, Le Guennec JY. Long-chain acylcarnitines regulate the hERG
pointes arrhythmias. J Cardiovasc Electrophysiol 21: 441–447, channel. PLoS One 7: e41686, 2012. doi:10.1371/journal.pone.
2010. doi:10.1111/j.1540-8167.2009.01641.x. 0041686.

1040. Roden DM, Abraham RL. Refining repolarization reserve. Heart 1053. Thorsen K, Dam VS, Kjaer-Sorensen K, Pedersen LN, Skeberdis
VA, Jurevicius J, Treinys R, Petersen IM, Nielsen MS, Oxvig C,
Rhythm 8: 1756–1757, 2011. doi:10.1016/j.hrthm.2011.06.024.
Morth JP, Matchkov VV, Aalkjaer C, Bundgaard H, Jensen HK.
1041. Roden DM, Yang T. Protecting the heart against arrhythmias: potas- Loss-of-activity-mutation in the cardiac chloride-bicarbonate
sium current physiology and repolarization reserve. Circulation 112: exchanger AE3 causes short QT syndrome. Nat Commun 8: 1696,
1376–1378, 2005. doi:10.1161/CIRCULATIONAHA.105.562777. 2017. doi:10.1038/s41467-017-01630-0.

1042. Antzelevitch C. Cardiac repolarization. The long and short of it. 1054. Nof E, Burashnikov A, Antzelevitch C. Cellular basis for atrial fibrilla-
Europace 7, Suppl 2: 3–9, 2005. doi:10.1016/j.eupc.2005.05.010. tion in an experimental model of short QT1: implications for a phar-
macological approach to therapy. Heart Rhythm 7: 251–257, 2010.
1043. Gussak I, Brugada P, Brugada J, Wright RS, Kopecky SL, Chaitman doi:10.1016/j.hrthm.2009.10.017.
BR, Bjerregaard P. Idiopathic short QT interval: a new clinical syn-
drome? Cardiology 94: 99–102, 2000. doi:10.1159/000047299. 1055. Shinnawi R, Shaheen N, Huber I, Shiti A, Arbel G, Gepstein A,
Ballan N, Setter N, Tijsen AJ, Borggrefe M, Gepstein L. Modeling
1044. Priori SG, Wilde AA, Horie M, Cho Y, Behr ER, Berul C, et al. reentry in the short QT syndrome with human-induced pluripotent
Executive summary: HRS/EHRA/APHRS expert consensus state- stem cell-derived cardiac cell sheets. J Am Coll Cardiol 73: 2310–
ment on the diagnosis and management of patients with inherited 2324, 2019. doi:10.1016/j.jacc.2019.02.055.

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1173


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

1056. Brugada P, Brugada J. Right bundle branch block, persistent ST 1068. Gandjbakhch E, Redheuil A, Pousset F, Charron P, Frank R. Clinical
segment elevation and sudden cardiac death: a distinct clinical and diagnosis, imaging, and genetics of arrhythmogenic right ventricu-
electrocardiographic syndrome. A multicenter report. J Am Coll lar cardiomyopathy/dysplasia: JACC State-of-the-Art Review. J Am
Cardiol 20: 1391–1396, 1992. doi:10.1016/0735-1097(92)90253-j. Coll Cardiol 72: 784–804, 2018. doi:10.1016/j.jacc.2018.05.065.

1057. Marcus FI, Fontaine GH, Guiraudon G, Frank R, Laurenceau JL, 1069. Que D, Yang P, Song X, Liu L. Traditional vs. genetic pathogenesis
Malergue C, Grosgogeat Y. Right ventricular dysplasia: a report of of arrhythmogenic right ventricular cardiomyopathy. Europace 17:
24 adult cases. Circulation 65: 384–398, 1982. doi:10.1161/01. 1770–1776, 2015. doi:10.1093/europace/euv042.
cir.65.2.384.
1070. Sen-Chowdhry S, Syrris P, McKenna WJ. Genetics of right ventricu-
1058. Kapplinger JD, Tester DJ, Alders M, Benito B, Berthet M, Brugada J, lar cardiomyopathy. J Cardiovasc Electrophysiol 16: 927–935,
Brugada P, Fressart V, Guerchicoff A, Harris-Kerr C, Kamakura S, 2005. doi:10.1111/j.1540-8167.2005.40842.x.
Kyndt F, Koopmann TT, Miyamoto Y, Pfeiffer R, Pollevick GD,
Probst V, Zumhagen S, Vatta M, Towbin JA, Shimizu W, Schulze- 1071. Sen-Chowdhry S, Lowe MD, Sporton SC, McKenna WJ.
Bahr E, Antzelevitch C, Salisbury BA, Guicheney P, Wilde AA, Arrhythmogenic right ventricular cardiomyopathy: clinical presenta-
Brugada R, Schott JJ, Ackerman MJ. An international compendium tion, diagnosis, and management. Am J Med 117: 685–695, 2004.
of mutations in the SCN5A-encoded cardiac sodium channel in doi:10.1016/j.amjmed.2004.04.028.
patients referred for Brugada syndrome genetic testing. Heart
1072. Corrado D, Basso C, Thiene G, McKenna WJ, Davies MJ, Fontaliran
Rhythm 7: 33–46, 2010. doi:10.1016/j.hrthm.2009.09.069.
F, Nava A, Silvestri F, Blomstrom-Lundqvist C, Wlodarska EK,
1059. Meregalli PG, Tan HL, Probst V, Koopmann TT, Tanck MW, Bhuiyan Fontaine G, Camerini F. Spectrum of clinicopathologic manifesta-
ZA, Sacher F, Kyndt F, Schott JJ, Albuisson J, Mabo P, Bezzina CR, tions of arrhythmogenic right ventricular cardiomyopathy/dysplasia:
Le Marec H, Wilde AA. Type of SCN5A mutation determines clinical a multicenter study. J Am Coll Cardiol 30: 1512–1520, 1997. doi:
severity and degree of conduction slowing in loss-of-function so- 10.1016/s0735-1097(97)00332-x.
dium channelopathies. Heart Rhythm 6: 341–348, 2009. doi:
10.1016/j.hrthm.2008.11.009. 1073. Maron BJ, Shirani J, Poliac LC, Mathenge R, Roberts WC, Mueller
FO. Sudden death in young competitive athletes. Clinical, demo-
1060. Tukkie R, Sogaard P, Vleugels J, de Groot IK, Wilde AA, Tan HL. graphic, and pathological profiles. JAMA 276: 199–204, 1996.
Delay in right ventricular activation contributes to Brugada syn-
drome. Circulation 109: 1272–1277, 2004. doi:10.1161/01.CIR.0000 1074. Corrado D, Basso C, Leoni L, Tokajuk B, Bauce B, Frigo G, Tarantini
118467.53182.D1. G, Napodano M, Turrini P, Ramondo A, Daliento L, Nava A, Buja G,
Iliceto S, Thiene G. Three-dimensional electroanatomic voltage
1061. Antzelevitch C. The Brugada syndrome: diagnostic criteria and cel- mapping increases accuracy of diagnosing arrhythmogenic right
lular mechanisms. Eur Heart J 22: 356–363, 2001. doi:10.1053/ ventricular cardiomyopathy/dysplasia. Circulation 111: 3042–3050,
euhj.2000.2461. 2005. doi:10.1161/CIRCULATIONAHA.104.486977.

1062. Antzelevitch C, Patocskai B. Brugada syndrome: clinical, genetic, 1075. Wijnmaalen AP, Schalij MJ, Bootsma M, Kies P, DE Roos A, Putter
molecular, cellular, and ionic aspects. Curr Probl Cardiol 41: 7–57, H, Bax JJ, Zeppenfeld K. Patients with scar-related right ventricular
2016. doi:10.1016/j.cpcardiol.2015.06.002. tachycardia: determinants of long-term outcome. J Cardiovasc
Electrophysiol 20: 1119–1127, 2009. doi:10.1111/j.1540-8167.2009.
1063. Verkerk AO, Remme CA, Schumacher CA, Scicluna BP, Wolswinkel 01516.x.
R, de Jonge B, Bezzina CR, Veldkamp MW. Functional Nav1.8 chan-
nels in intracardiac neurons: the link between SCN10A and cardiac 1076. Coumel P, Fidelle J, Lucet V, Attuel PY. Catecholamine induced
electrophysiology. Circ Res 111: 333–343, 2012. doi:10.1161/ severe ventricular arrhythmias with Adams-Stokes syndrome in
CIRCRESAHA.112.274035. children: report of four cases. Br Heart J : 28–37, 1978.

1064. Chambers JC, Zhao J, Terracciano CM, Bezzina CR, Zhang W, 1077. Liu N, Rizzi N, Boveri L, Priori SG. Ryanodine receptor and calse-
Kaba R, Navaratnarajah M, Lotlikar A, Sehmi JS, Kooner MK, Deng questrin in arrhythmogenesis: what we have learnt from genetic
G, Siedlecka U, Parasramka S, El-Hamamsy I, Wass MN, Dekker LR, diseases and transgenic mice. J Mol Cell Cardiol 46: 149–159,
de Jong JS, Sternberg MJ, McKenna W, Severs NJ, de Silva R, 2009. doi:10.1016/j.yjmcc.2008.10.012.
Wilde AA, Anand P, Yacoub M, Scott J, Elliott P, Wood JN, Kooner
JS. Genetic variation in SCN10A influences cardiac conduction. Nat 1078. Iyer V, Hajjar RJ, Armoundas AA. Mechanisms of abnormal calcium
Genet 42: 149–152, 2010. doi:10.1038/ng.516. homeostasis in mutations responsible for catecholaminergic poly-
morphic ventricular tachycardia. Circ Res 100: e22–e31, 2007.
1065. Bezzina CR, Barc J, Mizusawa Y, Remme CA, Gourraud JB, doi:10.1161/01.RES.0000258468.31815.42.
Simonet F, et al. Common variants at SCN5A-SCN10A and HEY2
are associated with Brugada syndrome, a rare disease with high 1079. Chiang DY, Kim JJ, Valdes SO, de la Uz C, Fan Y, Orcutt J, Domino
risk of sudden cardiac death. Nat Genet 45: 1044–1049, 2013. M, Smith M, Wehrens XH, Miyake CY. Loss-of-function SCN5A
doi:10.1038/ng.2712. mutations associated with sinus node dysfunction, atrial arrhyth-
mias, and poor pacemaker capture. Circ Arrhythm Electrophysiol
1066. Gaborit N, Wichter T, Varro A, Szuts V, Lamirault G, Eckardt L, Paul 8: 1105–1112, 2015. doi:10.1161/CIRCEP.115.003098.
M, Breithardt G, Schulze-Bahr E, Escande D, Nattel S, Demolombe
S. Transcriptional profiling of ion channel genes in Brugada syn- 1080. Fatkin D, Otway R, Vandenberg JI. Genes and atrial fibrillation: a
drome and other right ventricular arrhythmogenic diseases. Eur new look at an old problem. Circulation 116: 782–792, 2007.
Heart J 30: 487–496, 2009. doi:10.1093/eurheartj/ehn520. doi:10.1161/CIRCULATIONAHA.106.688889.

1067. Awad MM, Calkins H, Judge DP. Mechanisms of disease: molecular 1081. Ehdaie A, Cingolani E, Shehata M, Wang X, Curtis AB, Chugh SS.
genetics of arrhythmogenic right ventricular dysplasia/cardiomyop- Sex differences in cardiac arrhythmias: clinical and research impli-
athy. Nat Clin Pract Cardiovasc Med 5: 258–267, 2008. doi:10. cations. Circ Arrhythm Electrophysiol 11: e005680, 2018. doi:10.
1038/ncpcardio1182. 1161/CIRCEP.117.005680.

1174 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
ION CHANNELS, ACTION POTENTIALS, AND ARRHYTHMIAS

1082. Abi-Gerges N, Philp K, Pollard C, Wakefield I, Hammond TG, potassium currents in rabbits. J Pharmacol Exp Ther 285: 672–
Valentin JP. Sex differences in ventricular repolarization: from 679, 1998.
cardiac electrophysiology to Torsades de Pointes. Fundam Clin
Pharmacol 18: 139–151, 2004. doi:10.1111/j.1472-8206.2004. 1097. Kurokawa J, Tamagawa M, Harada N, Honda S, Bai CX, Nakaya H,
00230.x. Furukawa T. Acute effects of oestrogen on the guinea pig and
human IKr channels and drug-induced prolongation of cardiac
1083. Makkar RR, Fromm BS, Steinman RT, Meissner MD, Lehmann MH. repolarization. J Physiol 586: 2961–2973, 2008. doi:10.1113/
Female gender as a risk factor for torsades de pointes associated jphysiol.2007.150367.
with cardiovascular drugs. JAMA 270: 2590–2597, 1993. doi:10.
1001/jama.1993.03510210076031. 1098. Liu XK, Katchman A, Whitfield BH, Wan G, Janowski EM, Woosley
RL, Ebert SN. In vivo androgen treatment shortens the QT interval
1084. Merri M, Benhorin J, Alberti M, Locati E, Moss AJ. and increases the densities of inward and delayed rectifier potas-
Electrocardiographic quantitation of ventricular repolarization. sium currents in orchiectomized male rabbits. Cardiovasc Res 57:
Circulation 80: 1301–1308, 1989. doi:10.1161/01.cir.80.5.1301. 28–36, 2003. doi:10.1016/S0008-6363(02)00673-9.

1085. Wolbrette DL. Risk of proarrhythmia with class III antiarrhythmic 1099. Nakamura H, Kurokawa J, Bai CX, Asada K, Xu J, Oren RV, Zhu ZI,
agents: sex-based differences and other issues. Am J Cardiol 91: Clancy CE, Isobe M, Furukawa T. Progesterone regulates cardiac
39D–44D, 2003. doi:10.1016/s0002-9149(02)03378-7. repolarization through a nongenomic pathway: an in vitro patch-
clamp and computational modeling study. Circulation 116: 2913–
1086. Kundu P, Ciobotaru A, Foroughi S, Toro L, Stefani E, Eghbali M. 2922, 2007. doi:10.1161/CIRCULATIONAHA.107.702407.
Hormonal regulation of cardiac KCNE2 gene expression. Mol Cell
Endocrinol 292: 50–62, 2008. doi:10.1016/j.mce.2008.06.003. 1100. Odening KE, Koren G, Kirk M. Normalization of QT interval duration
in a long QT syndrome patient during pregnancy and the postpar-
1087. Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Sch€ utz G, tum period due to sex hormone effects on cardiac repolarization.
Umesono K, Blumberg B, Kastner P, Mark M, Chambon P, Evans Heart Rhythm Case Rep 2: 223–227, 2016. doi:10.1016/j.hrcr.
RM. The nuclear receptor superfamily: the second decade. Cell 83: 2015.12.012.
835–839, 1995. doi:10.1016/0092-8674(95)90199-x.
1101. Xiao L, Zhang L, Han W, Wang Z, Nattel S. Sex-based transmural
1088. Furukawa T, Kurokawa J. Regulation of cardiac ion channels via differences in cardiac repolarization and ionic-current properties in
non-genomic action of sex steroid hormones: implication for the canine left ventricles. Am J Physiol Heart Circ Physiol 291: H570–
gender difference in cardiac arrhythmias. Pharmacol Ther 115: H580, 2006. doi:10.1152/ajpheart.01288.2005.
106–115, 2007. doi:10.1016/j.pharmthera.2007.04.008.
1102. Abi-Gerges N, Small BG, Lawrence CL, Hammond TG, Valentin JP,
1089. Taneja T, Mahnert BW, Passman R, Goldberger J, Kadish A. Effects Pollard CE. Gender differences in the slow delayed (IKs) but not in
of sex and age on electrocardiographic and cardiac electrophysio- inward (IK1) rectifier K1 currents of canine Purkinje fibre cardiac
logical properties in adults. Pacing Clin Electrophysiol 24: 16–21, action potential: key roles for IKs, beta-adrenoceptor stimulation,
2001. doi:10.1046/j.1460-9592.2001.00016.x. pacing rate and gender. Br J Pharmacol 147: 653–660, 2006.
doi:10.1038/sj.bjp.0706491.
1090. El Khoury N, Mathieu S, Marger L, Ross J, El Gebeily G, Ethier N,
Fiset C. Upregulation of the hyperpolarization-activated current 1103. Lindenfeld J, Cleveland JC Jr, Kao DP, White M, Wichman S,
increases pacemaker activity of the sinoatrial node and heart rate Bristow JC, Peterson V, Rodegheri-Brito J, Korst A, Blain-Nelson P,
during pregnancy in mice. Circulation 127: 2009–2020, 2013. Sederberg J, Hunt SA, Gilbert EM, Ambardekar AV, Minobe W, Port
doi:10.1161/CIRCULATIONAHA.113.001689. JD, Bristow MR. Sex-related differences in age-associated down-
regulation of human ventricular myocardial beta1-adrenergic recep-
1091. Tsai WC, Chen YC, Kao YH, Lu YY, Chen SA, Chen YJ. Distinctive tors. J Heart Lung Transplant 35: 352–361, 2016. doi:10.1016/j.
sodium and calcium regulation associated with sex differences in healun.2015.10.021.
atrial electrophysiology of rabbits. Int J Cardiol 168: 4658–4666,
2013. doi:10.1016/j.ijcard.2013.07.183. 1104. Barajas-Martinez H, Haufe V, Chamberland C, Roy MJ, Fecteau
MH, Cordeiro JM, Dumaine R. Larger dispersion of INa in female
1092. Tsai WC, Chen YC, Lin YK, Chen SA, Chen YJ. Sex differences in dog ventricle as a mechanism for gender-specific incidence of car-
the electrophysiological characteristics of pulmonary veins and left diac arrhythmias. Cardiovasc Res 81: 82–89, 2009. doi:10.1093/
atrium and their clinical implication in atrial fibrillation. Circ cvr/cvn255.
Arrhythm Electrophysiol 4: 550–559, 2011. doi:10.1161/CIRCEP.
111.961995. 1105. Yang X, Chen G, Papp R, Defranco DB, Zeng F, Salama G.
Oestrogen upregulates L-type Ca21 channels via oestrogen-recep-
1093. Song Y, Shryock JC, Belardinelli L. An increase of late sodium cur- tor- by a regional genomic mechanism in female rabbit hearts. J
rent induces delayed afterdepolarizations and sustained triggered Physiol 590: 493–508, 2012. doi:10.1113/jphysiol.2011.219501.
activity in atrial myocytes. Am J Physiol Heart Circ Physiol 294:
H2031–H2039, 2008. doi:10.1152/ajpheart.01357.2007. 1106. Odening KE, Choi BR, Liu GX, Hartmann K, Ziv O, Chaves L,
Schofield L, Centracchio J, Zehender M, Peng X, Brunner M, Koren
1094. Bai CX, Kurokawa J, Tamagawa M, Nakaya H, Furukawa T. G. Estradiol promotes sudden cardiac death in transgenic long QT
Nontranscriptional regulation of cardiac repolarization currents by type 2 rabbits while progesterone is protective. Heart Rhythm 9:
testosterone. Circulation 112: 1701–1710, 2005. doi:10.1161/ 823–832, 2012. doi:10.1016/j.hrthm.2012.01.009.
CIRCULATIONAHA.104.523217.
1107. Er F, Michels G, Brandt MC, Khan I, Haase H, Eicks M, Lindner M,
1095. James AF, Arberry LA, Hancox JC. Gender-related differences in Hoppe UC. Impact of testosterone on cardiac L-type calcium chan-
ventricular myocyte repolarization in the guinea pig. Basic Res nels and Ca21 sparks: acute actions antagonize chronic effects.
Cardiol 99: 183–192, 2004. doi:10.1007/s00395-003-0451-6. Cell Calcium 41: 467–477, 2007. doi:10.1016/j.ceca.2006.09.003.

1096. Liu XK, Katchman A, Drici MD, Ebert SN, Ducic I, Morad M, Woosley 1108. Chen G, Yang X, Alber S, Shusterman V, Salama G. Regional
RL. Gender difference in the cycle length-dependent QT and genomic regulation of cardiac sodium-calcium exchanger by

Physiol Rev  VOL 101  JULY 2021  www.prv.org 1175


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.
VARRÓ ET AL.

oestrogen. J Physiol 589: 1061–1080, 2011. doi:10.1113/jphysiol. logical treatment and its modulation by genetic variation.
2010.203398. Neuropsychobiology 77: 67–72, 2019. doi:10.1159/000493400.

1109. Tsang S, Wong SS, Wu S, Kravtsov GM, Wong TM. Testosterone- 1123. Sicouri S, Antzelevitch C. Mechanisms underlying the actions of
augmented contractile responses to alpha1- and beta1-adrenocep- antidepressant and antipsychotic drugs that cause sudden cardiac
tor stimulation are associated with increased activities of RyR, arrest. Arrhythm Electrophysiol Rev 7: 199–209, 2018. doi:10.
SERCA, and NCX in the heart. Am J Physiol Cell Physiol 296: 15420/aer.2018.29.2.
C766–C782, 2009. doi:10.1152/ajpcell.00193.2008.
1124. Cardiac Arrhythmia Suppression Trial (CAST) Investigators.
1110. Yan S, Chen Y, Dong M, Song W, Belcher SM, Wang HS. Bisphenol Preliminary report: effect of encainide and flecainide on mortality in
A and 17beta-estradiol promote arrhythmia in the female heart via a randomized trial of arrhythmia suppression after myocardial
alteration of calcium handling. PLoS One 6: e25455, 2011. infarction. N Engl J Med 321: 406–412, 1989. doi:10.1056/
doi:10.1371/journal.pone.0025455. NEJM198908103210629.

1111. Lu YY, Cheng CC, Chen YC, Lin YK, Chen SA, Chen YJ. Electrolyte 1125. Siebels J, Kuck KH. Implantable cardioverter defibrillator compared
disturbances differentially regulate sinoatrial node and pulmonary with antiarrhythmic drug treatment in cardiac arrest survivors (the
vein electrical activity: A contribution to hypokalemia- or hyponatre- Cardiac Arrest Study Hamburg). Am Heart J 127: 1139–1144, 1994.
mia-induced atrial fibrillation. Heart Rhythm 13: 781–788, 2016. doi:10.1016/0002-8703(94)90101-5.
doi:10.1016/j.hrthm.2015.12.005.
1126. Waldo AL, Camm AJ, deRuyter H, Friedman PL, MacNeil DJ, Pauls
1112. Osadchii OE, Larsen AP, Olesen SP. Predictive value of electrical JF, Pitt B, Pratt CM, Schwartz PJ, Veltri EP. Effect of d-sotalol on
restitution in hypokalemia-induced ventricular arrhythmogenicity. mortality in patients with left ventricular dysfunction after recent
Am J Physiol Heart Circ Physiol 298: H210–H220, 2010. doi:10. and remote myocardial infarction. The SWORD Investigators.
1152/ajpheart.00695.2009. Survival With Oral d-Sotalol. Lancet 348: 7–12, 1996. doi:10.1016/
S0140-6736(96)02149-6.
1113. Tazmini K, Frisk M, Lewalle A, Laasmaa M, Morotti S, Lipsett DB,
Manfra O, Skogested J, Aronsen JM, Sejersted OM, Sjaastad I, 1127. Frommeyer G, Eckardt L. Drug-induced proarrhythmia: risk factors
Edwards AG, Grandi E, Niederer SA, Øie E, Louch WE. and electrophysiological mechanisms. Nat Rev Cardiol 13: 36–47,
Hypokalemia promotes arrhythmia by distinct mechanisms in atrial 2016. doi:10.1038/nrcardio.2015.110.
and ventricular myocytes. Circ Res 126: 889–906, 2020.
1128. Food and Drug Administration. International Conference on
doi:10.1161/CIRCRESAHA.119.315641.
Harmonisation; guidance on S7B Nonclinical Evaluation of the
1114. Shimoni Y, Clark RB, Giles WR. Role of an inwardly rectifying potas- Potential for Delayed Ventricular Repolarization (QT Interval
sium current in rabbit ventricular action potential. J Physiol 448: Prolongation) by Human Pharmaceuticals; availability. Notice. Fed
709–727, 1992. doi:10.1113/jphysiol.1992.sp019066. Regist 70: 61133–61134, 2005.

1115. Scamps F, Carmeliet E. Delayed K1 current and external K1 in sin- 1129. Nattel S, Yue L, Wang Z. Cardiac ultrarapid delayed rectifiers: a
gle cardiac Purkinje cells. Am J Physiol Cell Physiol 257: C1086– novel potassium current family of functional similarity and molecu-
C1092, 1989. doi:10.1152/ajpcell.1989.257.6.C1086. lar diversity. Cell Physiol Biochem 9: 217–226, 1999. doi:10.1159/
000016318.
1116. Eisner DA, Lederer WJ. The role of the sodium pump in the effects
of potassium-depleted solutions on mammalian cardiac muscle. J 1130. Ravens U, Wettwer E, Hálá O. Pharmacological modulation of ion
Physiol 294: 279–301, 1979. doi:10.1113/jphysiol.1979.sp012930. channels and transporters. Cell Calcium 35: 575–582, 2004.
doi:10.1016/j.ceca.2004.01.011.
1117. Osadchii OE. Mechanisms of hypokalemia-induced ventricular
arrhythmogenicity. Fundam Clin Pharmacol 24: 547–559, 2010. 1131. Spellmann I, Reinhard MA, Veverka D, Zill P, Obermeier M,
doi:10.1111/j.1472-8206.2010.00835.x. Dehning S, Schennach R, M€ €ller HJ, Riedel M, Musil R.
uller N, Mo
QTc prolongation in short-term treatment of schizophrenia patients:
1118. Bailie DS, Inoue H, Kaseda S, Ben-David J, Zipes DP. Magnesium effects of different antipsychotics and genetic factors. Eur Arch
suppression of early afterdepolarizations and ventricular tachyar- Psychiatry Clin Neurosci 268: 383–390, 2018. doi:10.1007/
rhythmias induced by cesium in dogs. Circulation 77: 1395–1402, s00406-018-0880-8.
1988. doi:10.1161/01.cir.77.6.1395.
1132. Woosley RL, Roden DM. Pharmacologic causes of arrhythmogenic
1119. Nagasawa S, Saitoh H, Kasahara S, Chiba F, Torimitsu S, Abe H, actions of antiarrhythmic drugs. Am J Cardiol 59: 19e–25e, 1987.
Yajima D, Iwase H. Relationship between KCNQ1 (LQT1) and doi:10.1016/0002-9149(87)90197-4.
KCNH2 (LQT2) gene mutations and sudden death during illegal
drug use. Sci Rep 8: 8443, 2018. doi:10.1038/s41598-018-26723-8. 1133. Hilgemann DW, Ball R. Regulation of cardiac Na1,Ca21 exchange
and KATP potassium channels by PIP2. Science 273: 956–959,
1120. Kaumann AJ. Adrenaline and noradrenaline increase contractile 1996. doi:10.1126/science.273.5277.956.
force of human ventricle through both beta 1- and beta 2-adreno-
ceptors. Biomed Biochim Acta 46: S411–S416, 1987. 1134. He Z, Feng S, Tong Q, Hilgemann DW, Philipson KD. Interaction of
PIP2 with the XIP region of the cardiac Na/Ca exchanger. Am J
1121. Silver PJ, Harris AL, Canniff PC, Lepore RE, Bentley RG, Hamel LT, Evans Physiol Cell Physiol 278: C661–C666, 2000. doi:10.1152/
DB. Phosphodiesterase isozyme inhibition, activation of the cAMP sys- ajpcell.2000.278.4.C661.
tem, and positive inotropy mediated by milrinone in isolated guinea pig
cardiac muscle. J Cardiovasc Pharmacol 13: 530–540, 1989. 1135. Cha TJ, Ehrlich JR, Zhang L, Nattel S, Wong TM. Atrial ionic remod-
eling induced by atrial tachycardia in the presence of congestive
1122. Corponi F, Fabbri C, Boriani G, Diemberger I, Albani D, Forloni G, heart failure. Circulation 110: 1520–1526, 2004. doi:10.1161/01.
Serretti A. Corrected QT interval prolongation in psychopharmaco- CIR.0000142052.03565.87.

1176 Physiol Rev  VOL 101  JULY 2021  www.prv.org


Downloaded from journals.physiology.org/journal/physrev (187.190.175.238) on November 6, 2023.

You might also like